Language selection

Search

Patent 2933568 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2933568
(54) English Title: MODIFIED NUCLEIC ACID MOLECULES AND USES THEREOF
(54) French Title: MOLECULES D'ACIDES NUCLEIQUES MODIFIES ET LEURS UTILISATIONS
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/85 (2006.01)
  • C7H 19/00 (2006.01)
  • C7H 21/02 (2006.01)
(72) Inventors :
  • ROY, ATANU (United States of America)
  • CONLEE, CHRISTOPHER R. (United States of America)
  • DE FOUGEROLLES, ANTONIN (Belgium)
  • FRALEY, ANDREW W. (United States of America)
(73) Owners :
  • MODERNA THERAPEUTICS, INC.
(71) Applicants :
  • MODERNA THERAPEUTICS, INC. (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2014-12-15
(87) Open to Public Inspection: 2015-06-18
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2014/070413
(87) International Publication Number: US2014070413
(85) National Entry: 2016-06-10

(30) Application Priority Data:
Application No. Country/Territory Date
61/916,052 (United States of America) 2013-12-13

Abstracts

English Abstract

The present disclosure provides modified nucleosides, nucleotides, and nucleic acids, and methods of using them.


French Abstract

La présente invention concerne des nucléosides, des nucléotides et des acides nucléiques modifiés, ainsi que leurs procédés d'utilisation.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. A polynucleotide, wherein at least two bases are 5-trifluoromethyl-cytosine
and 1-methyl-
pseudo-uracil; 5-hydroxymethyl-cytosine and 1-methyl-pseudo-uracil; 5-bromo-
cytosine and 1-methyl-
pseudo-uracil; 5-trifluoromethyl-cytosine and pseudo-uracil; 5-hydroxymethyl-
cytosine and pseudo-uracil;
5-bromo-cytosine and pseudo-uracil; cytosine and 5-methoxy-uracil; 5-methyl-
cytosine and 5-methoxy-
uracil; 5-trifluoromethyl-cytosine and 5-methoxy-uracil; 5-hydroxymethyl-
cytosine and 5-methoxy-uracil; or
5-bromo-cytosine and 5-methoxy-uracil.
2. The polynucleotide of claim 1, wherein at least two bases are 5-
trifluoromethyl-cytosine and 5-
methoxy-uracil; 5-hydroxymethyl-cytosine and 5-methoxy-uracil; or 5-bromo-
cytosine and 5-methoxy-
uracil.
3. The polynucleotide of claim 1, wherein at least two bases are 5-bromo-
cytosine and 5-
methoxy-uracil.
4. A polynucleotide, wherein at least one base is 1,6-Dimethyl-pseudo-uracil,
1-(optionally
substituted C1-C6Alkyl)-6-(1-propynyl)-pseudo-uracil, 1-(optionally
substituted C1-C6Alkyl)-6-(2-propynyl)-
pseudo-uracil, 1-(optionally substituted C1-C6Alkyl)-6-allyl-pseudo-uracil, 1-
(optionally substituted C1-C6
Alkyl)-6-ethynyl-pseudo-uracil, 1-(optionally substituted C1-C6Alkyl)-6-
homoallyl-pseudo-uracil, 1-
(optionally substituted C1-C6Alkyl)-6-vinyl-pseudo-uracil, 1-Methyl-6-(2,2,2-
Trifluoroethyl)pseudo-uracil,
1-Methyl-6-(4-morpholino)-pseudo-uracil, 1-Methyl-6-(4-thiomorpholino)-pseudo-
uracil, 1-Methyl-6-
(optionally substituted phenyl)pseudo-uracil, 1-Methyl-6-amino-pseudo-uracil,
1-Methyl-6-azido-pseudo-
uracil, 1-Methyl-6-bromo-pseudo-uracil, 1-Methyl-6-butyl-pseudo-uracil, 1-
Methyl-6-chloro-pseudo-uracil,
1-Methyl-6-cyano-pseudo-uracil, 1-Methyl-6-dimethylamino-pseudo-uracil, 1-
Methyl-6-ethoxy-pseudo-
uracil, 1-Methyl-6-ethylcarboxylate-pseudo-uracil, 1-Methyl-6-ethyl-pseudo-
uracil, 1-Methyl-6-fluoro-
pseudo-uracil, 1-Methyl-6-formyl-pseudo-uracil, 1-Methyl-6-hydroxyamino-pseudo-
uracil, 1-Methyl-6-
hydroxy-pseudo-uracil, 1-Methyl-6-iodo-pseudo-uracil, 1-Methyl-6-iso-propyl-
pseudo-uracil, 1-Methyl-6-
methoxy-pseudo-uracil, 1-Methyl-6-methylamino-pseudo-uracil, 1-Methyl-6-phenyl-
pseudo-uracil, 1-
Methyl-6-propyl-pseudo-uracil, 1-Methyl-6-tert-butyl-pseudo-uracil, 1-Methyl-6-
trifluoromethoxy-pseudo-
uracil, 1-Methyl-6-trifluoromethyl-pseudo-uracil, 6-(2,2,2-Trifluoroethyl)-
pseudo-uracil, 6-(4-Morpholino)-
pseudo-uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-(optionally substituted-
Phenyl)-pseudo-uracil, 6-
Amino-pseudo-uracil, 6-Azido-pseudo-uracil, 6-Bromo-pseudo-uracil, 6-Butyl-
pseudo-uracil, 6-Chloro-
pseudo-uracil, 6-Cyano-pseudo-uracil, 6-Dimethylamino-pseudo-uracil, 6-Ethoxy-
pseudo-uracil, 6-
Ethylcarboxylate-pseudo-uracil, 6-Ethyl-pseudo-uracil, 6-Fluoro-pseudo-uracil,
6-Formyl-pseudo-uracil, 6-
Hydroxyamino-pseudo-uracil, 6-Hydroxy-pseudo-uracil, 6-lodo-pseudo-uracil, 6-
iso-Propyl-pseudo-uracil,
6-Methoxy-pseudo-uracil, 6-Methylamino-pseudo-uracil, 6-Methyl-pseudo-uracil,
6-Phenyl-pseudo-uracil,
6-Propyl-pseudo-uracil, 6-tert-Butyl-pseudo-uracil, 6-Trifluoromethoxy-pseudo-
uracil, 6-Trifluoromethyl-
pseudo-uracil, 1-(3-Amino-3-carboxypropyl)pseudo-uracil, 1-(2,2,2-
Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-
Trimethyl-benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-phenyl)pseudo-uracil, 1-(2-
Amino-2-
carboxyethyl)pseudo-uracil, 1-(2-Amino-ethyl)pseudo-uracil, 1-(3-Amino-
propyl)pseudo-uracil, 1-(4-
Amino-4-carboxybutyl)pseudo-uracil, 1-(4-Amino-benzyl)pseudo-uracil, 1-(4-
Amino-butyl)pseudo-uracil,
- 347 -

1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-benzyl)pseudo-uracil, 1-(4-
Methoxy-phenyl)pseudo-
uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil,
1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-hexyl)pseudo-uracil, 1-Aminomethyl-
pseudo-uracil, 1-
Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-Cyclobutylmethyl-pseudo-uracil,
1-Cyclobutyl-pseudo-
uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-
uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-
Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-
Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-uracil, 1-Hexyl-pseudo-uracil, 1-iso-
Propyl-pseudo-uracil 1-
Pentyl-pseudo-uracil, 1-Phenyl-pseudo-uracil, 1-Propyl-pseudo-uracil, 1-p-
toluyl-pseudo-uracil, 1-tert-
Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-uracil, 3-(optionally
substituted C1-C6Alkyl)-pseudo-uracil,
Pseudo-uracil-N1-2-ethanoic acid, Pseudo-uracil-N1-3-propionic acid, Pseudo-
uracil-N1-4-butanoic acid,
Pseudo-uracil-N1-5-pentanoic acid, Pseudo-uracil-N1-6-hexanoic acid, Pseudo-
uracil-N1-7-heptanoic
acid, Pseudo-uracil-N1-methyl-p-benzoic acid, 6-phenyl-pseudo-uracil, 6-azido-
pseudo-uracil, Pseudo-
uracil-N1-p-benzoic acid, N3-Methyl-pseudo-uracil, 5-Methyl-amino-methyl-
uracil, 5-Carboxy-methyl-
amino-methyl-uracil, 5-(carboxyhydroxymethyl)uracil methyl ester 5-
(carboxyhydroxymethyl)uracil, 2-
anhydro-cytosine, 2-anhydro-uracil, 5-Methoxycarbonylmethyl-2-thio-uracil, 5-
Methylaminomethyl-2-
seleno-uracil, 5-(iso-Pentenylaminomethyl)-uracil, 5-(iso-Pentenylaminomethyl)-
2-thio-uracil, 5-(iso-
Pentenylaminomethyl)- uracil, 5-Trideuteromethyl-6-deutero-uracil, 5-(2-Chloro-
phenyl)-2-thio-cytosine, 5-
(4-Amino-phenyl)-2-thio-cytosine, 5-(2-Furanyl)-uracil, 8-Trifluoromethyl-
adenine, 2-Trifluoromethyl-
adenine, 3-Deaza-3-fluoro-adenine, 3-Deaza-3-bromo-adenine, 3-Deaza-3-iodo-
adenine, 1-
Hydroxymethyl-pseudo-uracil, 1-(2-Hydroxyethyl)-pseudo-uracil, 1-Methoxymethyl-
pseudo-uracil, 1-(2-
Methoxyethyl)-pseudo-uracil, 1-(2,2-Diethoxyethyl)-pseudo-uracil, 1-(2-
Hydroxypropyl)-pseudo-uracil,
(2R)-1-(2-Hydroxypropyl)-pseudo-uracil, (2S)-1-(2-Hydroxypropyl)-pseudo-
uracil, 1-Cyanomethyl-pseudo-
uracil, 1-Morpholinomethyl-pseudo-uracil, 1-Thiomorpholinomethyl-pseudo-
uracil, 1-Benzyloxymethyl-
pseudo-uracil, 1-(2,2,3,3,3-Pentafluoropropyl)-pseudo-uracil, 1-
Thiomethoxymethyl-pseudo-uracil, 1-
Methanesulfonylmethyl-pseudo-uracil, 1-Vinyl-pseudo-uracil, 1-Allyl-pseudo-
uracil, 1-Homoallyl-pseudo-
uracil, 1-Propargyl-pseudo-uracil, 1-(4-Fluorobenzyl)-pseudo-uracil, 1-(4-
Chlorobenzyl)-pseudo-uracil, 1-
(4-Bromobenzyl)-pseudo-uracil, 1-(4-lodobenzyl)-pseudo-uracil, 1-(4-
Methylbenzyl)-pseudo-uracil, 1-(4-
Trifluoromethylbenzyl)-pseudo-uracil, 1-(4-Methoxybenzyl)-pseudo-uracil, 1-(4-
Trifluoromethoxybenzyl)-
pseudo-uracil, 1-(4-Thiomethoxybenzyl)-pseudo-uracil, 1-(4-
Methanesulfonylbenzyl)-pseudo-uracil,
Pseudo-uracil 1-(4-methylbenzoic acid), Pseudo-uracil 1-(4-
methylbenzenesulfonic acid), 1-[2,4,6-
Trimethylbenzyl)-pseudo-uracil, 1-(4-Nitrobenzyl)-pseudo-uracil, 1-(4-
Azidobenzyl)-pseudo-uracil, 1-[3,4-
Dimethoxybenzyl)-pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyl)-pseudo-
uracil, 1-Acetyl-pseudo-
uracil, 1-Trifluoroacetyl-pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-
pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyl)-pseudo-uracil, Pseudo-uracil 1-methylphosphonic acid
diethyl ester, Pseudo-
uracil 1-methylphosphonic acid, Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid,
Pseudo-uracil 1-[3-{2-(2-
ethoxy)-ethoxy}] propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-
ethoxy}]propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-(2-ethoxy )-ethoxy]-ethoxy )-ethoxy}]propionic
acid, Pseudo-uracil 1-[3-{2-(2-
[2-{2(2-ethoxy )-ethoxy}-ethoxyl]-ethoxy )-ethoxy}]propionic acid, 1-{3-[2-(2-
Aminoethoxy)-ethoxy]-
propionyl } pseudo-uracil, 1-[3-(2-{2-[2-(2-Aminoethoxy)-ethoxy]-ethoxy}-
ethoxy)-propionyl]-pseudo-uracil,
1-Biotinyl-pseudo-uracil, 1-Biotinyl-PEG2-pseudo-uracil, 5-(C3-8 cycloalkyl)-
cytosine, 5-methyl-N6-acetyl--
- 348 -

cytosine, 5-(carboxymethyl)-N6-trifluoroacetyl-cytosine trifluoromethyl ester,
N6-propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-
propionyl)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-pseudo-isocytosine, 1-hydroxy-pseudo-isocytosine,
or 1-(2,2,2-trifluoroethyl)-
pseudo-uracil.
5. The polynucleotide of claim 4, wherein as least one base is 1,6-Dimethyl-
pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-6-(1-propynyl)-pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-6-
(2-propynyl)-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-allyl-
pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-6-ethynyl-pseudo-uracil, 1-(optionally substituted C1-
C6 Alkyl)-6-homoallyl-
pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-vinyl-pseudo-uracil, 1-
Methyl-6-(2,2,2-
Trifluoroethyl)pseudo-uracil, 1-Methyl-6-(4-morpholino)-pseudo-uracil, 1-
Methyl-6-(4-thiomorpholino)-
pseudo-uracil, 1-Methyl-6-(optionally substituted phenyl)pseudo-uracil, 1-
Methyl-6-amino-pseudo-uracil,
1-Methyl-6-azido-pseudo-uracil, 1-Methyl-6-bromo-pseudo-uracil, 1-Methyl-6-
butyl-pseudo-uracil, 1-
Methyl-6-chloro-pseudo-uracil, 1-Methyl-6-cyano-pseudo-uracil, 1-Methyl-6-
dimethylamino-pseudo-uracil,
1-Methyl-6-ethoxy-pseudo-uracil, 1-Methyl-6-ethylcarboxylate-pseudo-uracil, 1-
Methyl-6-ethyl-pseudo-
uracil, 1-Methyl-6-fluoro-pseudo-uracil, 1-Methyl-6-formyl-pseudo-uracil, 1-
Methyl-6-hydroxyamino-
pseudo-uracil, 1-Methyl-6-hydroxy-pseudo-uracil, 1-Methyl-6-iodo-pseudo-
uracil, 1-Methyl-6-iso-propyl-
pseudo-uracil, 1-Methyl-6-methoxy-pseudo-uracil, 1-Methyl-6-methylamino-pseudo-
uracil, 1-Methyl-6-
phenyl-pseudo-uracil, 1-Methyl-6-propyl-pseudo-uracil, 1-Methyl-6-tert-butyl-
pseudo-uracil, 1-Methyl-6-
trifluoromethoxy-pseudo-uracil, 1-Methyl-6-trifluoromethyl-pseudo-uracil, 6-
(2,2,2-Trifluoroethyl)-pseudo-
uracil, 6-(4-Morpholino)-pseudo-uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-
(Substituted-Phenyl)-
pseudo-uracil, 6-Amino-pseudo-uracil, 6-Azido-pseudo-uracil, 6-Bromo-pseudo-
uracil, 6-Butyl-pseudo-
uracil, 6-Chloro-pseudo-uracil, 6-Cyano-pseudo-uracil, 6-Dimethylamino-pseudo-
uracil, 6-Ethoxy-pseudo-
uracil, 6-Ethylcarboxylate-pseudo-uracil, 6-Ethyl-pseudo-uracil, 6-Fluoro-
pseudo-uracil, 6-Formyl-pseudo-
uracil, 6-Hydroxyamino-pseudo-uracil, 6-Hydroxy-pseudo-uracil, 6-Iodo-pseudo-
uracil, 6-iso-Propyl-
pseudo-uracil, 6-Methoxy-pseudo-uracil, 6-Methylamino-pseudo-uracil, 6-Methyl-
pseudo-uracil, 6-Phenyl-
pseudo-uracil, 6-Phenyl-pseudo-uracil, 6-Propyl-pseudo-uracil, 6-tert-Butyl-
pseudo-uracil, 6-
Trifluoromethoxy-pseudo-uracil, 6-Trifluoromethyl-pseudo-uracil, 1-(3-Amino-3-
carboxypropyl)pseudo-
uracil, 1-(2,2,2-Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-Trimethyl-
benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-
phenyl)pseudo-uracil, 1-(2-Amino-2-carboxyethyl)pseudo-uracil, 1-(2-Amino-
ethyl)pseudo-uracil, 1-(3-
Amino-propyl)pseudo-uracil, 1-(4-Amino-4-carboxybutyl)pseudo-uracil, 1-(4-
Amino-benzyl)pseudo-uracil,
1-(4-Amino-butyl)pseudo-uracil, 1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-
benzyl)pseudo-uracil, 1-
(4-Methoxy-phenyl)pseudo-uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-
benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil, 1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-
hexyl)pseudo-uracil, 1-
Aminomethyl-pseudo-uracil, 1-Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-
Cyclobutylmethyl-pseudo-
uracil, 1-Cyclobutyl-pseudo-uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-
Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-
pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-
pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-
uracil, 1-Hexyl-pseudo-
uracil, 1-iso-Propyl-pseudo-uracil, 1-Pentyl-pseudo-uracil, 1-Phenyl-pseudo-
uracil, 1-Propyl-pseudo-
uracil, 1-p-tolyl-pseudo-uracil, 1-tert-Butyl-pseudo-uracil, 1-Trifluoromethyl-
pseudo-uracil, 3-(optionally
-349-

substituted C1-C6 Alkyl)-pseudo-uracil, Pseudo-uracil-N1-2-ethanoic acid,
Pseudo-uracil-N1-3-propionic
acid, Pseudo-uracil-N1-4-butanoic acid, Pseudo-uracil-N1-5-pentanoic acid,
Pseudo-uracil-N1-6-hexanoic
acid, Pseudo-uracil-N1-7-heptanoic acid, Pseudo-uracil-N1-methyl-p-benzoic
acid, 6-phenyl-pseudo-
uracil, 6-azido-pseudo-uracil, or Pseudo-uracil-N1-p-benzoic acid.
6. The polynucleotide of claim 4, wherein at least one base is N3-Methyl-
pseudo-uracil, 5-Methyl-
amino-methyl-uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-
(carboxyhydroxymethyl)uracil methyl ester
or 5-(carboxyhydroxymethyl)uracil.
7. The polynucleotide of claim 4, wherein at least one base is 2-anhydro-
cytosine or 2-anhydro-
uracil.
8. The polynucleotide of claim 4, wherein at least one base is 5-
Methoxycarbonylmethyl-2-thio-
uracil, 5-Methylaminomethyl-2-seleno-uracil, 5-(iso-pentenylaminomethyl)-
uracil, 5-(iso-
pentenylaminomethyl)- 2-thio-uracil, or 5-(iso-Pentenylaminomethyl)- uracil.
9. The polynucleotide of claim 4, wherein at least one base is 5-
Trideuteromethyl-6-deutero-
uracil, 5-(2-Chloro-phenyl)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-
cytosine, 5-(2-Furanyl)-uracil, N4-
methyl-cytosine, 8-Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-
3-fluoro-adenine, 3-
Deaza-3-bromo-adenine, or 3-Deaza-3-iodo-adenine.
10. The polynucleotide of claim 4, wherein at least one base is 1-
Hydroxymethyl-pseudo-uracil, 1-
(2-Hydroxyethyl)-pseudo-uracil, 1-Methoxymethyl-pseudo-uracil, 1-(2-
Methoxyethyl)-pseudo-uracil, 1-
(2,2-Diethoxyethyl)-pseudo-uracil, 1-(2-Hydroxypropyl)-pseudo-uracil, (2R)-1-
(2-Hydroxypropyl)-pseudo-
uracil, (2S)-1-(2-Hydroxypropyl)-pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-
Morpholinomethyl-
pseudo-uracil, 1-Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-
uracil, 1-(2,2,3,3,3-
Pentafluoropropyl)-pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-
Methanesulfonylmethyl-pseudo-
uracil, 1-Vinyl-pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-
uracil, 1-Propargyl-pseudo-uracil,
1-(4-Fluorobenzyl)-pseudo-uracil, 1-(4-Chlorobenzyl)-pseudo-uracil, 1-(4-
Bromobenzyl)-pseudo-uracil, 1-
(4-Iodobenzyl)-pseudo-uracil, 1-(4-Methylbenzyl)-pseudo-uracil, 1-(4-
Trifluoromethylbenzyl)-pseudo-
uracil, 1-(4-Methoxybenzyl)-pseudo-uracil, 1-(4-Trifluoromethoxybenzyl)-pseudo-
uracil, 1-(4-
Thiomethoxybenzyl)-pseudo-uracil, 1-(4-Methanesulfonylbenzyl)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
Trimethylbenzyl)-pseudo-
uracil, 1-(4-Nitrobenzyl)-pseudo-uracil, 1-(4-Azidobenzyl)-pseudo-uracil, 1-
(3,4-Dimethoxybenzyl)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyl)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyl)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 1-[3-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxy]-ethoxy
)-ethoxy}]propionic acid, 1-{3-[2-(2-Aminoethoxy)-ethoxy]-propionyl } pseudo-
uracil, 1-[3-(2-{2-[2-(2-
-350-

Aminoethoxy)-ethoxy]-ethoxy}-ethoxy)-propionyl]-pseudo-uracil, 1-Biotinyl-
pseudo-uracil, or 1-Biotinyl-
PEG2-pseudo-uracil.
11. The polynucleotide of claim 4, wherein at least one base is 5-cyclopropyl-
cytosine, 5-methyl-
N6-acetyl-cytosine, 5-(carboxymethyl)-N6-trifluoroacetyl-cytosine
trifluoromethyl ester, N6-propionyl-
cytosine, 5-monofluoromethyl-cytosine, 5-trifluoromethoxy-cytosine, N6-(1,1,1-
trifluoro-propionyl)-
cytosine, 4-acetyl-pseudo-isocytosine, 1-ethyl-pseudo-isocytosine, or 1-
hydroxy-pseudo-isocytosine.
12. The polynucleotide of claim 4, wherein at least one base is 1-(2,2,2-
trifluoroethyl)-pseudo-
uracil.
13. The polynucleotide of any one of claims 1-12, comprising at least one
backbone moiety of
Formula Vlll-Xll:
<IMG>
wherein the dashed line represents an optional double bond;
B is a nucleobase;
each of U and U' is, independently, O, S, N(R U)nu, or C(R U)nu, wherein nu is
an integer from 0 to 2
and each R U is, independently, H, halo, or optionally substituted C1-C6
alkyl;
each of R1', R2', R1", R2", R1, R3', R4, R5, R6, and R7 is, independently, H,
halo, hydroxy, thiol,
optionally substituted C1-C6alkyl, optionally substituted C2-C6alkynyl,
optionally substituted C1-C6
heteroalkyl, optionally substituted C2-C6heteroalkenyl, optionally substituted
C2-C6heteroalkynyl,
optionally substituted amino, azido, optionally substituted C6-C10 aryl; or R5
can join together with one or
more of R1', R1", R2', or R2" to form optionally substituted C1-C6alkylene or
optionally substituted C1-C6
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted C3-10 carbocycle or an optionally substituted C3-C9heterocyclyl;
or R4 can join together with
- 351 -

one or more of R1', R1", R2', R2", R3, or R5 to form optionally substituted C1-
C6 alkylene or optionally
substituted C1-C6 heteroalkylene and, taken together with the carbons to which
they are attached, provide
an optionally substituted C3-10 carbocycle or an optionally substituted C3-C9
heterocyclyl;
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6 heteroalkynyl, optionally substituted amino, azido, optionally
substituted C6-C10 aryl; or R3 can join
together with one or more of R1', R1", R2', R2", and, taken together with the
carbons to which they are
attached, provide an optionally substituted C3-10 carbocycle or an optionally
substituted C3-C9 heterocyclyl;
wherein if said optional double bond is present, R3 is absent;
each of m' and m" is, independently, an integer from 0 to 3;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, O, S, Se, -NR N1-,
optionally substituted C1-C6
alkylene, or optionally substituted C1-C6 heteroalkylene, wherein R N1 is H,
optionally substituted C1-C6
alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6
alkynyl, optionally substituted C6-
C10 aryl, or absent;
each Y4 is, independently, H, hydroxyl, protected hydroxyl, halo, thiol,
boranyl, optionally
substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl, optionally
substituted C2-C6 alkynyl, optionally
substituted C1-C6 heteroalkyl, optionally substituted C2-C6 heteroalkenyl,
optionally substituted C2-C6
heteroalkynyl, optionally substituted amino, or absent; and
Y5 is O, S, Se, optionally substituted C1-C6 alkylene, or optionally
substituted C1-C6
heteroalkylene.
14. The polynucleotide of any one of claims 1-13, further comprising:
(a) a 5' UTR optionally comprising at least one Kozak sequence;
(b) a 3' UTR; and
(c) at least one 5' cap structure.
15. The polynucleotide of claim 14, wherein at least one 5' cap structure is
Cap0, Cap1, ARCA,
inosine, N1-methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-
guanosine, 2-amino-
guanosine, LNA-guanosine, or 2-azido-guanosine.
16. The polynucleotide of any one of claims 1-15, further comprising a poly-A
tail.
17. The polynucleotide of any one of claims 14-16, wherein said polynucleotide
encodes a protein
of interest.
18. The polynucleotide of any one of claims 1-17, which is purified.
19. The polynucleotide of any one of claims 1-18, wherein said polynucleotide
is codon
optimized.
-352-

20. An isolated polynucleotide encoding a polypeptide of interest, said
isolated polynucleotide
comprising:
(a) a 5' UTR, e.g., comprising at least one Kozak sequence;
(b) a 3' UTR; and
(c) at least one 5' cap structure,
wherein at least one base is 1-Methyl-3-(3-amino-3-carboxypropyl)pseudo-
uracil, 5-Oxyacetic
acid-methyl ester-uracil, 5-Trifluoromethyl-cytosine, 5-Trifluoromethyl-
uracil, 5-
Carboxymethylaminomethyl-2-thio-uracil, 5-Methylaminomethyl-2-thio-uracil, 5-
Methoxy-carbonyl-methyl-
uracil, 5-Oxyacetic acid-uracil, 3-(3-Amino-3-carboxypropyl)-uracil, 2-Amino-
adenine, 8-Aza-adenine,
Xanthosine, 5-Bromo-cytosine, 5-Aminoallyl-cytosine, 5-iodo-cytosine, 8-bromo-
adenine, 8-bromo-
guanine, N4-Benzoyl-cytosine, N4-Amino-cytosine, N6-Bz-adenine, N2-isobutyl-
guanine, 5-
Methylaminomethyl-2-thio-uracil, 5-Carbamoylmethyl-uracil, 1-Methyl-3-(3-amino-
3-carboxypropyl)
pseudo-uracil, 5-Methyldihydro-uracil, 5-(1-propynyl)cytosine, 5-
Ethynylcytosine, 5-vinyl-uracil, (Z)-5-(2-
Bromo-vinyl)-uracil, (E)-5-(2-Bromo-vinyl)-uracil, 5-Methoxy-cytosine, 5-
Formyl-uracil, 5-Cyano-uracil, 5-
Dimethylamino-uracil, 5-Cyano-cytosine, 5-Phenylethynyl-uracil, (E)-5-(2-Bromo-
vinyl)-cytosine, 2-
Mercapto-adenine, 2-Azido-adenine, 2-Fluoro-adenine, 2-Chloro-adenine, 2-Bromo-
adenine, 2-Iodo-
adenine, 7-Amino-1H-pyrazolo[4,3-d]pyrimidine, 2,4-dihydropyrazolo[4,3-
d]pyrimidin-7-one, 2,4-
dihydropyrazolo[4,3-d]pyrimidine-5,7-dione, pyrrolosine, 9-Deaza-adenine, 9-
Deaza-guanine, 3-Deaza-
adenine, 3-Deaza-3-chloro-adenine, 1-Deaza-adenine, 5-vinyl-cytosine, 5-phenyl-
cytosine, 5-
difluoromethyl-cytosine, 5-(1-propynyl)-uracil, 5-(1-propynyl)-cytosine, or 5-
methoxymethyl-cytosine.
21. The polynucleotide of claim 20, wherein at least one base is 1-Methyl-3-(3-
amino-3-
carboxypropyl)pseudo-uracil.
22. The polynucleotide of claim 20, wherein at least one base is 5-Oxyacetic
acid-methyl ester-
uracil, 5-Trifluoromethyl-cytosine, 5-Trifluoromethyl-uracil, 5-
Carboxymethylaminomethyl-2-thio-uracil, 5-
Methylaminomethyl-2-thio-uracil, 5-Methoxy-carbonyl-methyl-uracil, 5-Oxyacetic
acid-uracil, or 3-(3-
Amino-3-carboxypropyl)-uracil.
23. The polynucleotide of claim 20, wherein at least one base is 2-Amino-
adenine, 8-Aza-
adenine, Xanthosine, 5-Bromo-cytosine, or 5-Aminoallyl-cytosine.
24. The polynucleotide of claim 20, wherein at least one base is 5-iodo-
cytosine, 8-bromo-
adenine, 8-bromo-guanine, N4-Benzoyl-cytosine, N4-Amino-cytosine, N6-Bz-
adenine, or N2-isobutyl-
guanine.
25. The polynucleotide of claim 20, wherein at least one base is 5-
Methylaminomethyl-2-thio-
uracil, 5-Carbamoylmethyl-uracil, 1-Methyl-3-(3-amino-3-carboxypropyl) pseudo-
uracil, or 5-
Methyldihydro-uracil.
-353-

26. The polynucleotide of claim 20, wherein at least one base is 5-(1-
propynyl)cytosine, 5-
Ethynylcytosine, 5-vinyl-uracil, (Z)-5-(2-Bromo-vinyl)-uracil, (E)-5-(2-Bromo-
vinyl)-uracil, 5-Methoxy-
cytosine, 5-Formyl-uracil, 5-Cyano-uracil, 5-Dimethylamino-uracil, 5-Cyano-
cytosine, 5-Phenylethynyl-
uracil, (E)-5-(2-Bromo-vinyl)-cytosine, 2-Mercapto-adenine, 2-Azido-adenine, 2-
Fluoro-adenine, 2-Chloro-
adenine, 2-Bromo-adenine, 2-lodo-adenine, 7-Amino-1H-pyrazolo[4,3-
d]pyrimidine, 2,4-
dihydropyrazolo[4,3-d]pyrimidin-7-one, 2,4-dihydropyrazolo[4,3-d]pyrimidine-
5,7-dione, pyrrolosine, 9-
Deaza-adenine, 9-Deaza-guanine, 3-Deaza-adenine, 3-Deaza-3-chloro-adenine, or
1-Deaza-adenine.
27. The polynucleotide of claim 20, wherein at least one base is 5-methoxy-
uracil, 5-vinyl-
cytosine, 5-phenyl-cytosine, 5-difluoromethyl-cytosine, or 5-methoxymethyl-
cytosine.
28. The polynucleotide of claim 20, wherein at least one base is 5-bromo-
cytosine.
29. The polynucleotide of any one of claims 20-28, further comprising a poly-A
tail.
30. The polynucleotide of any one of 20-29, wherein said polynucleotide is
purified.
31. The polynucleotide of any one of claims 20-30, wherein said at least one
5' cap structure is
Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-
guanosine, 8-oxo-
guanosine, 2-amino-guanosine, LNA-guanosine, or 2-azido-guanosine.
32. The polynucleotide of any one of claims 20-31, wherein said polynucleotide
is codon
optimized.
33. A compound of Formula I:
A-B,
Formula I
wherein A is:
<IMG>
- 354 -

<IMG>
wherein the dashed line represents an optional double bond;
each of U and U' is, independently, O, S, N(R U)nu, or C(R U)nu, wherein nu is
an integer from 0 to 2
and each R U is, independently, H, halo, or optionally substituted C1-C6
alkyl;
each of R1' , R2' , R1", R1, R3 , R4, R5, R6, and R7 is, independently, H,
halo, hydroxy, thiol, optionally
substituted C1-C6 alkyl, optionally substituted C2-C6 alkynyl, optionally
substituted C1-C6 heteroalkyl,
optionally substituted C2-C6 heteroalkenyl, optionally substituted C2-C6
heteroalkynyl, optionally
substituted amino, azido, optionally substituted C6-C10 aryl; or R5 can join
together with one or more of R1',
R1 , R2 , or R2 to form optionally substituted C1-C6 alkylene or optionally
substituted C1-C6 heteroalkylene
and, taken together with the carbons to which they are attached, provide an
optionally substituted C3-10
carbocycle or an optionally substituted C3-C9 heterocyclyl; or R4 can join
together with one or more of R1 ,
R1 , R2 , R2 , R3, or R5 to form optionally substituted C1-C6 alkylene or
optionally substituted C1-C6
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted C3-10 carbocycle or an optionally substituted C3-C9 heterocyclyl;
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6 heteroalkynyl, optionally substituted amino, azido, optionally
substituted C6-C10 aryl; or R3 can join
together with one or more of R1', R1", R2' , R2" , and, taken together with
the carbons to which they are
attached, provide an optionally substituted C3-10 carbocycle or an optionally
substituted C2-C9 heterocyclyl;
wherein if said optional double bond is present, R3 is absent;
each of m' and m" is, independently, an integer from 0 to 3;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, O, S, Se, -NR N1-,
optionally substituted C1-C6
alkylene, or optionally substituted C1-C6 heteroalkylene, wherein R N1 is H,
optionally substituted C1-C6
alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6
alkynyl, optionally substituted C6-
C10 aryl, or absent;
each of Y4 and Y6 is, independently, H, hydroxyl, protected hydroxyl, halo,
thiol, boranyl,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl,
optionally substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6 heteroalkynyl, optionally substituted amino, or absent;
Y5 is O, S, Se, optionally substituted C1-C6 alkylene, or optionally
substituted C1-C6
heteroalkylene; and
B is 1 ,6-Dimethyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-(1-
propynyl)-pseudo-
uracil, 1-(optionally substituted C1-C6 Alkyl)-6-(2-propynyl)-pseudo-uracil, 1-
(optionally substituted C1-C6
- 355 -

Alkyl)-6-allyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-ethynyl-
pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-6-homoallyl-pseudo-uracil, 1-(optionally substituted
C1-C6 Alkyl)-6-vinyl-pseudo-
uracil, 1-Methyl-6-(2,2,2-Trifluoroethyl)pseudo-uracil, 1-Methyl-6-(4-
morpholino)-pseudo-uracil, 1-Methyl-
6-(4-thiomorpholino)-pseudo-uracil, 1-Methyl-6-(optionally substituted
phenyl)pseudo-uracil, 1-Methyl-6-
amino-pseudo-uracil, 1-Methyl-6-azido-pseudo-uracil, 1-Methyl-6-bromo-pseudo-
uracil, 1-Methyl-6-butyl-
pseudo-uracil, 1-Methyl-6-chloro-pseudo-uracil, 1-Methyl-6-cyano-pseudo-
uracil, 1-Methyl-6-
dimethylamino-pseudo-uracil, 1-Methyl-6-ethoxy-pseudo-uracil, 1-Methyl-6-
ethylcarboxylate-pseudo-
uracil, 1-Methyl-6-ethyl-pseudo-uracil, 1-Methyl-6-fluoro-pseudo-uracil, 1-
Methyl-6-formyl-pseudo-uracil,
1-Methyl-6-hydroxyamino-pseudo-uracil, 1-Methyl-6-hydroxy-pseudo-uracil, 1-
Methyl-6-iodo-pseudo-
uracil, 1-Methyl-6-iso-propyl-pseudo-uracil, 1-Methyl-6-methoxy-pseudo-uracil,
1-Methyl-6-methylamino-
pseudo-uracil, 1-Methyl-6-phenyl-pseudo-uracil, 1-Methyl-6-propyl-pseudo-
uracil, 1-Methyl-6-tert-butyl-
pseudo-uracil, 1-Methyl-6-trifluoromethoxy-pseudo-uracil, 1-Methyl-6-
trifluoromethyl-pseudo-uracil, 6-
(2,2,2-Trifluoroethyl)-pseudo-uracil, 6-(4-Morpholino)-pseudo-uracil, 6-(4-
Thiomorpholino)-pseudo-uracil,
6-(optionally substituted-Phenyl)-pseudo-uracil, 6-Amino-pseudo-uracil, 6-
Azido-pseudo-uracil, 6-Bromo-
pseudo-uracil, 6-Butyl-pseudo-uracil, 6-Chloro-pseudo-uracil, 6-Cyano-pseudo-
uracil, 6-Dimethylamino-
pseudo-uracil, 6-Ethoxy-pseudo-uracil, 6-Ethylcarboxylate-pseudo-uracil, 6-
Ethyl-pseudo-uracil, 6-Fluoro-
pseudo-uracil, 6-Formyl-pseudo-uracil, 6-Hydroxyamino-pseudo-uracil, 6-Hydroxy-
pseudo-uracil, 6-lodo-
pseudo-uracil, 6-iso-Propyl-pseudo-uracil, 6-Methoxy-pseudo-uracil, 6-
Methylamino-pseudo-uracil, 6-
Methyl-pseudo-uracil, 6-Phenyl-pseudo-uracil, 6-Propyl-pseudo-uracil, 6-tert-
Butyl-pseudo-uracil, 6-
Trifluoromethoxy-pseudo-uracil, 6-Trifluoromethyl-pseudo-uracil, 1-(3-Amino-3-
carboxypropyl)pseudo-
uracil, 1-(2,2,2-Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-Trimethyl-
benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-
phenyl)pseudo-uracil, 1-(2-Amino-2-carboxyethyl)pseudo-uracil, 1-(2-Amino-
ethyl)pseudo-uracil, 1-(3-
Amino-propyl)pseudo-uracil, 1-(4-Amino-4-carboxybutyl)pseudo-uracil, 1-(4-
Amino-benzyl)pseudo-uracil,
1-(4-Amino-butyl)pseudo-uracil, 1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-
benzyl)pseudo-uracil, 1-
(4-Methoxy-phenyl)pseudo-uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-
benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil, 1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-
hexyl)pseudo-uracil, 1-
Aminomethyl-pseudo-uracil, 1-Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-
Cyclobutylmethyl-pseudo-
uracil, 1-Cyclobutyl-pseudo-uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-
Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-
pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-
pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-
uracil, 1-Hexyl-pseudo-
uracil, 1-iso-Propyl-pseudo-uracil 1-Pentyl-pseudo-uracil, 1-Phenyl-pseudo-
uracil, 1-Propyl-pseudo-uracil,
1-p-toluyl-pseudo-uracil, 1-tert-Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-
uracil, 3-(optionally
substituted C1-C6 Alkyl)-pseudo-uracil, Pseudo-uracil-N1-2-ethanoic acid,
Pseudo-uracil-N1-3-propionic
acid, Pseudo-uracil-N1-4-butanoic acid, Pseudo-uracil-N1-5-pentanoic acid,
Pseudo-uracil-N1-6-hexanoic
acid, Pseudo-uracil-N1-7-heptanoic acid, Pseudo-uracil-N1-methyl-p-benzoic
acid, 6-phenyl-pseudo-
uracil, 6-azido-pseudo-uracil, Pseudo-uracil-N1-p-benzoic acid, N3-Methyl-
pseudo-uracil, 5-Methyl-
amino-methyl-uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-
(carboxyhydroxymethyl)uracil methyl ester
5-(carboxyhydroxymethyl)uracil, 2-anhydro-cytosine, 2-anhydro-uracil, 5-
Methoxycarbonylmethyl-2-thio-
uracil, 5-Methylaminomethyl-2-seleno-uracil, 5-(iso-Pentenylaminomethyl)-
uracil, 5-(iso-
Pentenylaminomethyl)- 2-thio-uracil, 5-(iso-Pentenylaminomethyl)- uracil, 5-
Trideuteromethyl-6-deutero-
- 356 -

uracil, 5-(2-Chloro-phenyl)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-
cytosine, 5-(2-Furanyl)-uracil, 8-
Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine,
3-Deaza-3-bromo-
adenine, 3-Deaza-3-iodo-adenine, 1-Hydroxymethyl-pseudo-uracil, 1-(2-
Hydroxyethyl)-pseudo-uracil, 1-
Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-pseudo-uracil, 1-(2,2-
Diethoxyethyl)-pseudo-uracil, 1-
(2-Hydroxypropyl)-pseudo-uracil, (2R)-1-(2-Hydroxypropyl)-pseudo-uracil, (2S)-
1-(2-Hydroxypropyl)-
pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-Morpholinomethyl-pseudo-uracil,
1-
Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil, 1-
(2,2,3,3,3-Pentafluoropropyl)-
pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-Methanesulfonylmethyl-
pseudo-uracil, 1-Vinyl-
pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-uracil, 1-Propargyl-
pseudo-uracil, 1-(4-
Fluorobenzyl)-pseudo-uracil, 1-(4-Chlorobenzyl)-pseudo-uracil, 1-(4-
Bromobenzyl)-pseudo-uracil, 1-(4-
lodobenzyl)-pseudo-uracil, 1-(4-Methylbenzyl)-pseudo-uracil, 1-(4-
Trifluoromethylbenzyl)-pseudo-uracil,
1-(4-Methoxybenzyl)-pseudo-uracil, 1-(4-Trifluoromethoxybenzyl)-pseudo-uracil,
1-(4-
Thiomethoxybenzyl)-pseudo-uracil, 1-(4-Methanesulfonylbenzyl)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
Trimethylbenzyl)-pseudo-
uracil, 1-(4-Nitrobenzyl)-pseudo-uracil, 1-(4-Azidobenzyl)-pseudo-uracil, 1-
(3,4-DiMethoxybenzyl)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyl)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyl)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 1-[3-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxy]-ethoxy
)-ethoxy}]propionic acid, 1-{3-[2-(2-Aminoethoxy)-ethoxy]-propionyl } pseudo-
uracil, 1-[3-(2-{2-[2-(2-
Aminoethoxy)-ethoxy]-ethoxy}-ethoxy)-propionyl]-pseudo-uracil, 1-Biotinyl-
pseudo-uracil, 1-Biotinyl-
PEG2-pseudo-uracil, 5-cyclopropyl-cytosine, 5-methyl-N6-acetyl- -cytosine, 5-
(carboxymethyl)-N6-
trifluoroacetyl-cytosine trifluoromethyl ester, N6-propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-
trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-propionyl)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-
pseudo-isocytosine, 1-hydroxy-pseudo-isocytosine, or 1-(2,2,2-trifluoroethyl)-
pseudo-uracil;
or a salt thereof.
34. The compound of claim 33, wherein B is 1,6-Dimethyl-pseudo-uracil, 1-
(optionally substituted
C1-C6 Alkyl)-6-(1-propynyl)-pseudo-uracil, 1-(optionally substituted C1-C6
Alkyl)-6-(2-propynyl)-pseudo-
uracil, 1-(optionally substituted C1-C6 Alkyl)-6-allyl-pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-6-
ethynyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-homoallyl-
pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-6-vinyl-pseudo-uracil, 1-Methyl-6-(2,2,2-
Trifluoroethyl)pseudo-uracil, 1-Methyl-6-
(4-morpholino)-pseudo-uracil, 1-Methyl-6-(4-thiomorpholino)-pseudo-uracil, 1-
Methyl-6-(optionally
substituted phenyl)pseudo-uracil, 1-Methyl-6-amino-pseudo-uracil, 1-Methyl-6-
azido-pseudo-uracil, 1-
Methyl-6-bromo-pseudo-uracil, 1-Methyl-6-butyl-pseudo-uracil, 1-Methyl-6-
chloro-pseudo-uracil, 1-
Methyl-6-cyano-pseudo-uracil, 1-Methyl-6-dimethylamino-pseudo-uracil, 1-Methyl-
6-ethoxy-pseudo-uracil,
1-Methyl-6-ethylcarboxylate-pseudo-uracil, 1-Methyl-6-ethyl-pseudo-uracil, 1-
Methyl-6-fluoro-pseudo-
uracil, 1-Methyl-6-formyl-pseudo-uracil, 1-Methyl-6-hydroxyamino-pseudo-
uracil, 1-Methyl-6-hydroxy-
pseudo-uracil, 1-Methyl-6-iodo-pseudo-uracil, 1-Methyl-6-iso-propyl-pseudo-
uracil, 1-Methyl-6-methoxy-
- 357 -

pseudo-uracil, 1-Methyl-6-methylamino-pseudo-uracil, 1-Methyl-6-phenyl-pseudo-
uracil, 1-Methyl-6-
propyl-pseudo-uracil, 1-Methyl-6-tert-butyl-pseudo-uracil, 1-Methyl-6-
trifluoromethoxy-pseudo-uracil, 1-
Methyl-6-trifluoromethyl-pseudo-uracil, 6-(2,2,2-Trifluoroethyl)-pseudo-
uracil, 6-(4-Morpholino)-pseudo-
uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-(Substituted-Phenyl)-pseudo-
uracil, 6-Amino-pseudo-uracil,
6-Azido-pseudo-uracil, 6-Bromo-pseudo-uracil, 6-Butyl-pseudo-uracil, 6-Chloro-
pseudo-uracil, 6-Cyano-
pseudo-uracil, 6-Dimethylamino-pseudo-uracil, 6-Ethoxy-pseudo-uracil, 6-
Ethylcarboxylate-pseudo-uracil,
6-Ethyl-pseudo-uracil, 6-Fluoro-pseudo-uracil, 6-Formyl-pseudo-uracil, 6-
Hydroxyamino-pseudo-uracil, 6-
Hydroxy-pseudo-uracil, 6-Iodo-pseudo-uracil, 6-iso-Propyl-pseudo-uracil, 6-
Methoxy-pseudo-uracil, 6-
Methylamino-pseudo-uracil, 6-Methyl-pseudo-uracil, 6-Phenyl-pseudo-uracil, 6-
Phenyl-pseudo-uracil, 6-
Propyl-pseudo-uracil, 6-tert-Butyl-pseudo-uracil, 6-Trifluoromethoxy-pseudo-
uracil, 6-Trifluoromethyl-
pseudo-uracil, 1-(3-Amino-3-carboxypropyl)pseudo-uracil, 1-(2,2,2-
Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-
Trimethyl-benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-phenyl)pseudo-uracil, 1-(2-
Amino-2-
carboxyethyl)pseudo-uracil, 1-(2-Amino-ethyl)pseudo-uracil, 1-(3-Amino-
propyl)pseudo-uracil, 1-(4-
Amino-4-carboxybutyl)pseudo-uracil, 1-(4-Amino-benzyl)pseudo-uracil, 1-(4-
Amino-butyl)pseudo-uracil,
1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-benzyl)pseudo-uracil, 1-(4-
Methoxy-phenyl)pseudo-
uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil,
1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-hexyl)pseudo-uracil, 1-Aminomethyl-
pseudo-uracil, 1-
Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-Cyclobutylmethyl-pseudo-uracil,
1-Cyclobutyl-pseudo-
uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-
uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-
Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-
Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-uracil, 1-Hexyl-pseudo-uracil, 1-iso-
Propyl-pseudo-uracil, 1-
Pentyl-pseudo-uracil, 1-Phenyl-pseudo-uracil, 1-Propyl-pseudo-uracil, 1-p-
tolyl-pseudo-uracil, 1-tert-
Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-uracil, 3-(optionally
substituted C1-C6 Alkyl)-pseudo-uracil,
Pseudo-uracil-N1-2-ethanoic acid, Pseudo-uracil-N1-3-propionic acid, Pseudo-
uracil-N1-4-butanoic acid,
Pseudo-uracil-N1-5-pentanoic acid, Pseudo-uracil-N1-6-hexanoic acid, Pseudo-
uracil-N1-7-heptanoic
acid, Pseudo-uracil-N1-methyl-p-benzoic acid, 6-phenyl-pseudo-uracil, 6-azido-
pseudo-uracil, or Pseudo-
uracil-N1-p-benzoic acid.
35. The compound of claim 33, wherein B is N3-Methyl-pseudo-uracil, 5-Methyl-
amino-methyl-
uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-(carboxyhydroxymethyl)uracil
methyl ester or 5-
(carboxyhydroxymethyl)uracil.
36. The compound of claim 33, wherein B is 2-anhydro-cytosine or 2-anhydro-
uracil.
37. The compound of claim 33, wherein B is 5-Methoxycarbonylmethyl-2-thio-
uracil, 5-
Methylaminomethyl-2-seleno-uracil, 5-(iso-Pentenylaminomethyl)-uracil, 5-(iso-
Pentenylaminomethyl)-2-
thio-uracil, or 5-(iso-Pentenylaminomethyl)-uracil.
38. The compound of claim 33, wherein B is 5-Trideuteromethyl-6-deutero-
uracil, 5-(2-Chloro-
phenyl)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-cytosine, 5-(2-Furanyl)-
uracil, N4-methyl-cytosine, 8-
-358-

Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine,
3-Deaza-3-bromo-
adenine, or 3-Deaza-3-iodo-adenine.
39. The compound of claim 33, wherein B is 1-Hydroxymethyl-pseudo-uracil, 1-(2-
Hydroxyethyl)-
pseudo-uracil, 1-Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-pseudo-
uracil, 1-(2,2-Diethoxyethyl)-
pseudo-uracil, (~)1-(2-Hydroxypropyl)-pseudo-uracil, (2R)-1-(2-Hydroxypropyl)-
pseudo-uracil, (2S)-1-(2-
Hydroxypropyl)-pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-Morpholinomethyl-
pseudo-uracil, 1-
Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil, 1-
(2,2,3,3,3-Pentafluoropropyl)-
pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-Methanesulfonylmethyl-
pseudo-uracil, 1-Vinyl-
pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-uracil, 1-Propargyl-
pseudo-uracil, 1-(4-
Fluorobenzyl)-pseudo-uracil, 1-(4-Chlorobenzyl)-pseudo-uracil, 1-(4-
Bromobenzyl)-pseudo-uracil, 1-(4-
Iodobenzyl)-pseudo-uracil, 1-(4-Methylbenzyl)-pseudo-uracil, 1-(4-
Trifluoromethylbenzyl)-pseudo-uracil,
1-(4-Methoxybenzyl)-pseudo-uracil, 1-(4-Trifluoromethoxybenzyl)-pseudo-uracil,
1-(4-
Thiomethoxybenzyl)-pseudo-uracil, 1-(4-Methanesulfonylbenzyl)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
Trimethylbenzyl)-pseudo-
uracil, 1-(4-Nitrobenzyl)-pseudo-uracil, 1-(4-Azidobenzyl)-pseudo-uracil, 1-
(3,4-Dimethoxybenzyl)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyl)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyl)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 1-[3-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxy]-ethoxy
)-ethoxy}]propionic acid, 1-{3-[2-(2-Aminoethoxy)-ethoxy]-propionyl } pseudo-
uracil, 1-[3-(2-{2-[2-(2-
Aminoethoxy)-ethoxy]-ethoxy}-ethoxy)-propionyl]-pseudo-uracil, 1-Biotinyl-
pseudo-uracil, or 1-Biotinyl-
PEG2-pseudo-uracil.
40. The compound of claim 33, wherein B is 5-cyclopropyl-cytosine, 5-methyl-N6-
acetyl-cytosine,
5-(carboxymethyl)-N6-trifluoroacetyl-cytosine trifluoromethyl ester, N6-
propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-
propionyl)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-pseudo-isocytosine, or 1-hydroxy-pseudo-
isocytosine.
41. The compound of claim 40, wherein B is 1-(2,2,2-trifluoroethyl)-
pseudouracil.
42. The compound of any one of claims 33-41, wherein A has the structure of
Formula II.
43. The compound of claim 42, wherein m' is 0.
44. The compound of claim 42 or 43, wherein m" is 1.
45. The compound of any one of claims 42-44, wherein R4 is hydrogen.
-359-

46. The compound of any one of claims 42-45, wherein A is:
<IMG>
U is O, S, N(R U)nu, or C(R U)nu, wherein nu is an integer from 1 to 2 and
each R U is, independently,
H, halo, or optionally substituted C1-C6 alkyl;
each of R1", R2", and R5 is, independently, H, halo, hydroxy, thiol,
optionally substituted C1-C6
alkyl, optionally substituted C2-C6 alkynyl, optionally substituted C1-C6
heteroalkyl, optionally substituted
C2-C6 heteroalkenyl, optionally substituted C2-C6 heteroalkynyl, optionally
substituted amino, azido,
optionally substituted C6-C10 aryl; or R5 can join together with one of R1 or
R2 to form optionally
substituted C1-C6 alkylene or optionally substituted C1-C6 heteroalkylene and,
taken together with the
carbons to which they are attached, provide an optionally substituted C3-10
carbocycle or an optionally
substituted C3-C9 heterocyclyl; or;
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6 heteroalkynyl, optionally substituted amino, azido, optionally
substituted C6-C10 aryl; or R3 can join
together with one of R1" or R2", and, taken together with the carbons to which
they are attached, provide
an optionally substituted C3-10 carbocycle or an optionally substituted C3-C9
heterocyclyl;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, O, S, Se, -NR N1-,
optionally substituted C1-C6
alkylene, or optionally substituted C1-C6 heteroalkylene, wherein R N1 is H,
optionally substituted C1-C6
alkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6
alkynyl, optionally substituted C6-
C10 aryl, or absent; and
each of Y4 and Y6 is, independently, H, hydroxyl, protected hydroxyl, halo,
thiol, boranyl,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6 alkenyl,
optionally substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6 heteroalkynyl, optionally substituted amino, or absent; and
Y5 is O, S, Se, optionally substituted C1-C6 alkylene, or optionally
substituted C1-C6
heteroalkylene.
47. The compound of any one of claims 42-44, wherein R2" is hydroxyl.
48. The compound of claim 47, wherein R1" is hydrogen.
49. The compound of claim 47 or 48, wherein R3 is hydrogen, and R5 is
hydrogen.
-360-

50. The compound of claim 47 or 48, wherein R3 is hydrogen, and R5 is
optionally substituted C2-
C6 alkynyl.
51. The compound of claim 50, wherein said optionally substituted C2-C6
alkynyl is ethynyl.
52. The compound of claim 47 or 48, wherein R5 is hydrogen.
53. The compound of claim 52, wherein R3 is azido or optionally substituted C2-
C6 alkynyl.
54. The compound of claim 53, wherein R3 is azido.
55. The compound of claim 53, wherein R3 is optionally substituted C2-C6
alkynyl, wherein said
optionally substituted C2-C6 alkynyl is ethynyl.
56. The compound of claim 47, wherein R3 is hydrogen and R5 is hydrogen.
57. The compound of claim 56, wherein R1" is optionally substituted C1-C6
alkyl or optionally
substituted C2-C6 alkynyl.
58. The compound of claim 57, wherein R1" is optionally substituted C1-C6
alkyl, wherein said
optionally substituted C1-C6 alkyl is trifluoromethyl.
59. The compound of claim 57, wherein R1" is optionally substituted C2-C6
alkynyl, wherein said
optionally substituted C2-C6 alkynyl is ethynyl.
60. The compound of any one of claims 42-46, wherein R2" is hydrogen.
61. The compound of claim 60, wherein R3 is hydrogen.
62. The compound of claim 60 or 61, wherein R5 is hydrogen.
63. The compound of any one of claims 60-62, wherein R1" is halo, thiol,
optionally substituted C1-
C6 heteroalkyl, azido, or amino.
64. The compound of claim 63, wherein said halo is fluoro, chloro, bromo, or
iodo.
65. The compound of claim 63, wherein said optionally substituted C1-C6
heteroalkyl is
thiomethoxy.
66. The compound of any one of claims 42-46, wherein R3 is hydrogen.
-361-

67. The compound of claim 66, wherein R5 is hydrogen.
68. The compound of claim 66 or 67, wherein R1" is hydroxy.
69. The compound of claim 68, wherein R2" is hydrogen, optionally substituted
C1-C6 alkyl, or
optionally substituted C2-C6 alkynyl.
70. The compound of claim 69, wherein said optionally substituted C1-C6 alkyl
is trifluoromethyl.
71. The compound of claim 69, wherein said optionally substituted C2-C6
alkynyl is ethynyl.
72. The compound of claim 66 or 67, wherein R1" is hydrogen.
73. The compound of claim 72, wherein R2" is thiol, optionally substituted C1-
C6 heteroalkyl, azido,
or amino.
74. The compound of claim 73, wherein said optionally substituted C1-C6
heteroalkyl is
thiomethoxy.
75. The compound of claim 66 or 67, wherein R1" is halo.
76. The compound of claim 75, wherein said halo is fluoro.
77. The compound of claim 75 or 76, wherein R2" is halo.
78. The compound of claim 77, wherein said halo is fluoro.
79. The compound of any one of claim 42-78, wherein U is C(R U)nu.
80. The compound of claim 79, wherein nu is 2.
81. The compound of claim 79 or 80, wherein each R u is hydrogen.
82. The compound of any one of claims 42-81, wherein q is 0; and Y6 is
hydroxyl.
83. The compound of any one of claims 42-82, wherein R5 is hydroxyl.
84. The compound of any one of claims 42-83, wherein Y5 is optionally
substituted C1-C6
alkylene.
-362-

85. The compound of claim 84, wherein said optionally substituted C1-C6
alkylene is methylene.
86. The compound of any one of claims 42-85, wherein r is 0, and Y6 is
hydroxyl.
87. The compound of any one of claims 42-85, wherein r is 3; each Y1 and Y3 is
O; and Y4 and Y6
are hydroxyl.
88. The compound of any one of claim 42-85, wherein r is 3, each Y1 is O; and
Y4 and Y6 are
hydroxyl.
89. The compound of claim 88, wherein at least one Y3 is S.
- 363 -

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
MODIFIED NUCLEIC ACID MOLECULES AND USES THEREOF
TECHNICAL FIELD
The present disclosure provides compositions and methods using modified
nucleic acids
to modulate cellular function. The modified nucleic acids of the invention may
encode peptides,
polypeptides or multiple proteins. The encoded molecules may be used as
therapeutics and/or
diagnostics.
BACKGROUND OF THE INVENTION
There are multiple problems with prior methodologies of effecting protein
expression. For
example, heterologous DNA introduced into a cell can be inherited by daughter
cells (whether or not
the heterologous DNA has integrated into the chromosome) or by offspring.
Introduced DNA can
integrate into host cell genomic DNA at some frequency, resulting in
alterations and/or damage to the
host cell genomic DNA. In addition, multiple steps must occur before a protein
is made. Once inside
the cell, DNA must be transported into the nucleus where it is transcribed
into RNA. The RNA
transcribed from DNA must then enter the cytoplasm where it is translated into
protein. This need for
multiple processing steps creates lag times before the generation of a protein
of interest. Further, it is
difficult to obtain DNA expression in cells; frequently DNA enters cells but
is not expressed or not
expressed at reasonable rates or concentrations. This can be a particular
problem when DNA is
introduced into cells such as primary cells or modified cell lines.
Naturally occurring RNAs are synthesized from four basic ribonucleotides: ATP,
CTP, UTP
and GTP, but may contain post-transcriptionally modified nucleotides. Further,
approximately one
hundred different nucleoside modifications have been identified in RNA
(Rozenski, J, Crain, P, and
McCloskey, J. (1999). The RNA Modification Database: 1999 update. Nucl Acids
Res 27: 196-197).
There is a need in the art for biological modalities to address the modulation
of
intracellular translation of nucleic acids. The present invention solves this
problem by providing new
mRNA molecules incorporating chemical modifications which impart properties
which are
advantageous to therapeutic development.
SUMMARY OF THE INVENTION
The present disclosure provides, inter alia, modified nucleosides, modified
nucleotides,
and modified nucleic acids whereby the modification is on the nucleobase,
sugar or backbone.
In a first aspect, the invention features a polynucleotide, wherein at least
two bases are 5-
trifluoromethyl-cytosine and 1-methyl-pseudo-uracil; 5-hydroxymethyl-cytosine
and 1-methyl-pseudo-
uracil; 5-bromo-cytosine and 1-methyl-pseudo-uracil; 5-trifluoromethyl-
cytosine and pseudo-uracil; 5-
hydroxymethyl-cytosine and pseudo-uracil; 5-bromo-cytosine and pseudo-uracil;
cytosine and 5-methoxy-
uracil; 5-methyl-cytosine and 5-methoxy-uracil; 5-trifluoromethyl-cytosine and
5-methoxy-uracil; 5-
hydroxymethyl-cytosine and 5-methoxy-uracil; or 5-bromo-cytosine and 5-methoxy-
uracil.
In some embodiments, at least two bases are 5-trifluoromethyl-cytosine and 5-
methoxy-uracil;
5-hydroxymethyl-cytosine and 5-methoxy-uracil; or 5-bromo-cytosine and 5-
methoxy-uracil.
In other embodiments, at least two bases are 5-bromo-cytosine and 5-methoxy-
uracil.
- 1 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In a second aspect, the invention features a polynucleotide, wherein at least
one base is 1,6-
Dimethyl-pseudo-uracil, 1-(optionally substituted C1-C6Alkyl)-6-(1-propyny1)-
pseudo-uracil, 1-(optionally
substituted C1-C6Alkyl)-6-(2-propyny1)-pseudo-uracil, 1-(optionally
substituted C1-C6Alkyl)-6-allyl-pseudo-
uracil, 1-(optionally substituted C1-C6Alkyl)-6-ethynyl-pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-
6-homoallyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-vinyl-
pseudo-uracil, 1-Methy1-6-(2,2,2-
Trifluoroethyl)pseudo-uracil, 1-Methy1-6-(4-morpholino)-pseudo-uracil, 1-
Methy1-6-(4-thiomorpholino)-
pseudo-uracil, 1-Methyl-6-(optionally substituted phenyl)pseudo-uracil, 1-
Methyl-6-amino-pseudo-uracil,
1-Methy1-6-azido-pseudo-uracil, 1-Methy1-6-bromo-pseudo-uracil, 1-Methy1-6-
butyl-pseudo-uracil, 1-
Methy1-6-chloro-pseudo-uracil, 1-Methy1-6-cyano-pseudo-uracil, 1-Methy1-6-
dimethylamino-pseudo-uracil,
1-Methy1-6-ethoxy-pseudo-uracil, 1-Methy1-6-ethylcarboxylate-pseudo-uracil, 1-
Methy1-6-ethyl-pseudo-
uracil, 1-Methy1-6-fluoro-pseudo-uracil, 1-Methy1-6-formyl-pseudo-uracil, 1-
Methy1-6-hydroxyamino-
pseudo-uracil, 1-Methyl-6-hydroxy-pseudo-uracil, 1-Methyl-6-iodo-pseudo-
uracil, 1-Methy1-6-iso-propyl-
pseudo-uracil, 1-Methy1-6-methoxy-pseudo-uracil, 1-Methy1-6-methylamino-pseudo-
uracil, 1-Methy1-6-
phenyl-pseudo-uracil, 1-Methy1-6-propyl-pseudo-uracil, 1-Methy1-6-tert-butyl-
pseudo-uracil, 1-Methy1-6-
trifluoromethoxy-pseudo-uracil, 1-Methy1-6-trifluoromethyl-pseudo-uracil, 6-
(2,2,2-Trifluoroethyl)-pseudo-
uracil, 6-(4-Morpholino)-pseudo-uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-
(optionally substituted-
Phenyl)-pseudo-uracil, 6-Amino-pseudo-uracil, 6-Azido-pseudo-uracil, 6-Bromo-
pseudo-uracil, 6-Butyl-
pseudo-uracil, 6-Chloro-pseudo-uracil, 6-Cyano-pseudo-uracil, 6-Dimethylamino-
pseudo-uracil, 6-Ethoxy-
pseudo-uracil, 6-Ethylcarboxylate-pseudo-uracil, 6-Ethyl-pseudo-uracil, 6-
Fluoro-pseudo-uracil, 6-Formyl-
pseudo-uracil, 6-Hydroxyamino-pseudo-uracil, 6-Hydroxy-pseudo-uracil, 6-lodo-
pseudo-uracil, 6-iso-
Propyl-pseudo-uracil, 6-Methoxy-pseudo-uracil, 6-Methylamino-pseudo-uracil, 6-
Methyl-pseudo-uracil, 6-
Phenyl-pseudo-uracil, 6-Propyl-pseudo-uracil, 6-tert-Butyl-pseudo-uracil, 6-
Trifluoromethoxy-pseudo-
uracil, 6-Trifluoromethyl-pseudo-uracil, 1-(3-Amino-3-carboxypropyl)pseudo-
uracil, 142,2,2-
Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-Trimethyl-benzyl)pseudo-uracil, 1-
(2,4,6-Trimethyl-phenyl)pseudo-
uracil, 1-(2-Amino-2-carboxyethyl)pseudo-uracil, 1-(2-Amino-ethyl)pseudo-
uracil, 1-(3-Amino-
propyl)pseudo-uracil, 1-(4-Amino-4-carboxybutyl)pseudo-uracil, 1-(4-Amino-
benzyl)pseudo-uracil, 1-(4-
Amino-butyl)pseudo-uracil, 1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-
benzyl)pseudo-uracil, 1-(4-
Methoxy-phenyl)pseudo-uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-
benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil, 1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-
hexyl)pseudo-uracil, 1-
Aminomethyl-pseudo-uracil, 1-Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-
Cyclobutylmethyl-pseudo-
uracil, 1-Cyclobutyl-pseudo-uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-
Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-
pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-
pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-
uracil, 1-Hexyl-pseudo-
uracil, 1-iso-Propyl-pseudo-uracil 1-Pentyl-pseudo-uracil, 1-Phenyl-pseudo-
uracil, 1-Propyl-pseudo-uracil,
1-p-toluyl-pseudo-uracil, 1-tert-Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-
uracil, 3-(optionally
substituted C1-C6 Alkyl)-pseudo-uracil, Pseudo-uracil-N1-2-ethanoic acid,
Pseudo-uracil-N1-3-propionic
acid, Pseudo-uracil-N1-4-butanoic acid, Pseudo-uracil-N1-5-pentanoic acid,
Pseudo-uracil-N1-6-hexanoic
acid, Pseudo-uracil-N1-7-heptanoic acid, Pseudo-uracil-N1-methyl-p-benzoic
acid, 6-phenyl-pseudo-
uracil, 6-azido-pseudo-uracil, Pseudo-uracil-N1-p-benzoic acid, N3-Methyl-
pseudo-uracil, 5-Methyl-
amino-methyl-uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-
(carboxyhydroxymethyl)uracil methyl ester
- 2 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
5-(carboxyhydroxymethyl)uracil, 2-anhydro-cytosine, 2-anhydro-uracil, 5-
Methoxycarbonylmethy1-2-thio-
uracil, 5-Methylaminomethy1-2-seleno-uracil, 5-(iso-Pentenylaminomethyl)-
uracil, 5-(iso-
Pentenylaminomethyl)- 2-thio-uracil, 5-(iso-Pentenylaminomethyl)- uracil, 5-
Trideuteromethy1-6-deutero-
uracil, 5-(2-Chloro-phenyl)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-
cytosine, 5-(2-FuranyI)-uracil, 8-
Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine,
3-Deaza-3-bromo-
adenine, 3-Deaza-3-iodo-adenine, 1-Hydroxymethyl-pseudo-uracil, 1-(2-
Hydroxyethyl)-pseudo-uracil, 1-
Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-pseudo-uracil, 1-(2,2-
Diethoxyethyl)-pseudo-uracil, 1-
(2-HydroxypropyI)-pseudo-uracil, (2R)-1-(2-HydroxypropyI)-pseudo-uracil, (2S)-
1-(2-HydroxypropyI)-
pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-Morpholinomethyl-pseudo-uracil,
1-
Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil, 1-
(2,2,3,3,3-PentafluoropropyI)-
pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-Methanesulfonylmethyl-
pseudo-uracil, 1-Vinyl-
pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-uracil, 1-Propargyl-
pseudo-uracil, 1-(4-
Fluorobenzy1)-pseudo-uracil, 1-(4-ChlorobenzyI)-pseudo-uracil, 1-(4-
BromobenzyI)-pseudo-uracil, 1-(4-
lodobenzyI)-pseudo-uracil, 1-(4-MethylbenzyI)-pseudo-uracil, 1-(4-
TrifluoromethylbenzyI)-pseudo-uracil,
1-(4-MethoxybenzyI)-pseudo-uracil, 1-(4-TrifluoromethoxybenzyI)-pseudo-uracil,
1-(4-
Thiomethoxybenzy1)-pseudo-uracil, 1-(4-MethanesulfonylbenzyI)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
TrimethylbenzyI)-pseudo-
uracil, 1-(4-NitrobenzyI)-pseudo-uracil, 1-(4-AzidobenzyI)-pseudo-uracil, 1-
(3,4-DimethoxybenzyI)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyI)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyI)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 143-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxyyethoxy
)-ethoxy}]propionic acid, 1-{342-(2-Aminoethoxy)-ethoxyypropionyl } pseudo-
uracil, 143-(2-{242-(2-
Aminoethoxy)-ethoxyyethoxy}-ethoxy)-propionylypseudo-uracil, 1-Biotinyl-pseudo-
uracil, 1-Biotinyl-
PEG2-pseudo-uracil, 5-(C3_8 cycloalkyl)-cytosine, 5-methyl-N6-acetyl--
cytosine, 5-(carboxymethyl)-N6-
trifluoroacetyl-cytosine trifluoromethyl ester, N6-propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-
trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-propionyI)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-
pseudo-isocytosine, 1-hydroxy-pseudo-isocytosine, or 1-(2,2,2-trifluoroethyl)-
pseudo-uracil.
In some embodiments, at least one base is 1,6-Dimethyl-pseudo-uracil, 1-
(optionally
substituted C1-C6 Alkyl)-6-(1-propyny1)-pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-6-(2-propyny1)-
pseudo-uracil, 1-(optionally substituted C1-C6Alkyl)-6-allyl-pseudo-uracil, 1-
(optionally substituted C1-C6
Alkyl)-6-ethynyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-
homoallyl-pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-6-vinyl-pseudo-uracil, 1-Methy1-6-(2,2,2-
TrifluoroethyDpseudo-uracil,
1-Methy1-6-(4-morpholino)-pseudo-uracil, 1-Methy1-6-(4-thiomorpholino)-pseudo-
uracil, 1-Methy1-6-
(optionally substituted phenyl)pseudo-uracil, 1-Methy1-6-amino-pseudo-uracil,
1-Methy1-6-azido-pseudo-
uracil, 1-Methy1-6-bromo-pseudo-uracil, 1-Methy1-6-butyl-pseudo-uracil, 1-
Methy1-6-chloro-pseudo-uracil,
1-Methy1-6-cyano-pseudo-uracil, 1-Methy1-6-dimethylamino-pseudo-uracil, 1-
Methy1-6-ethoxy-pseudo-
uracil, 1-Methy1-6-ethylcarboxylate-pseudo-uracil, 1-Methy1-6-ethyl-pseudo-
uracil, 1-Methy1-6-fluoro-
pseudo-uracil, 1-Methy1-6-formyl-pseudo-uracil, 1-Methy1-6-hydroxyamino-pseudo-
uracil, 1-Methy1-6-
- 3 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
hydroxy-pseudo-uracil, 1-Methyl-6-iodo-pseudo-uracil, 1-Methyl-6-iso-propyl-
pseudo-uracil, 1-Methy1-6-
methoxy-pseudo-uracil, 1-Methy1-6-methylamino-pseudo-uracil, 1-Methy1-6-phenyl-
pseudo-uracil, 1-
Methy1-6-propyl-pseudo-uracil, 1-Methy1-6-tert-butyl-pseudo-uracil, 1-Methy1-6-
trifluoromethoxy-pseudo-
uracil, 1-Methy1-6-trifluoromethyl-pseudo-uracil, 6-(2,2,2-Trifluoroethyl)-
pseudo-uracil, 6-(4-Morpholino)-
pseudo-uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-(Substituted-Phenyl)-
pseudo-uracil, 6-Amino-
pseudo-uracil, 6-Azido-pseudo-uracil, 6-Bromo-pseudo-uracil, 6-Butyl-pseudo-
uracil, 6-Chloro-pseudo-
uracil, 6-Cyano-pseudo-uracil, 6-Dimethylamino-pseudo-uracil, 6-Ethoxy-pseudo-
uracil, 6-
Ethylcarboxylate-pseudo-uracil, 6-Ethyl-pseudo-uracil, 6-Fluoro-pseudo-uracil,
6-Formyl-pseudo-uracil, 6-
Hydroxyamino-pseudo-uracil, 6-Hydroxy-pseudo-uracil, 6-lodo-pseudo-uracil, 6-
iso-Propyl-pseudo-uracil,
6-Methoxy-pseudo-uracil, 6-Methylamino-pseudo-uracil, 6-Methyl-pseudo-uracil,
6-Phenyl-pseudo-uracil,
6-Phenyl-pseudo-uracil, 6-Propyl-pseudo-uracil, 6-tert-Butyl-pseudo-uracil, 6-
Trifluoromethoxy-pseudo-
uracil, 6-Trifluoromethyl-pseudo-uracil, 1-(3-Amino-3-carboxypropyl)pseudo-
uracil, 142,2,2-
Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-Trimethyl-benzyl)pseudo-uracil, 1-
(2,4,6-Trimethyl-phenyl)pseudo-
uracil, 1-(2-Amino-2-carboxyethyl)pseudo-uracil, 1-(2-Amino-ethyl)pseudo-
uracil, 1-(3-Amino-
propyl)pseudo-uracil, 1-(4-Amino-4-carboxybutyl)pseudo-uracil, 1-(4-Amino-
benzyl)pseudo-uracil, 1-(4-
Amino-butyl)pseudo-uracil, 1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-
benzyl)pseudo-uracil, 1-(4-
Methoxy-phenyl)pseudo-uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-
benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil, 1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-
hexyl)pseudo-uracil, 1-
Aminomethyl-pseudo-uracil, 1-Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-
Cyclobutylmethyl-pseudo-
uracil, 1-Cyclobutyl-pseudo-uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-
Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-
pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-
pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-
uracil, 1-Hexyl-pseudo-
uracil, 1-iso-Propyl-pseudo-uracil, 1-Pentyl-pseudo-uracil, 1-Phenyl-pseudo-
uracil, 1-Propyl-pseudo-
uracil, 1-p-tolyl-pseudo-uracil, 1-tert-Butyl-pseudo-uracil, 1-Trifluoromethyl-
pseudo-uracil, 3-(optionally
substituted C1-C6 Alkyl)-pseudo-uracil, Pseudo-uracil-N1-2-ethanoic acid,
Pseudo-uracil-N1-3-propionic
acid, Pseudo-uracil-N1-4-butanoic acid, Pseudo-uracil-N1-5-pentanoic acid,
Pseudo-uracil-N1-6-hexanoic
acid, Pseudo-uracil-N1-7-heptanoic acid, Pseudo-uracil-N1-methyl-p-benzoic
acid, 6-phenyl-pseudo-
uracil, 6-azido-pseudo-uracil, or Pseudo-uracil-N1-p-benzoic acid.
In other embodiments, at least one base is N3-Methyl-pseudo-uracil, 5-Methyl-
amino-methyl-
uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-(carboxyhydroxymethyOuracil
methyl ester, or 5-
(carboxyhydroxymethyl)uracil.
In certain embodiments, at least one base is 2-anhydro-cytidine or 2-anhydro-
uracil.
In some embodiments, at least one base is 5-Methoxycarbonylmethy1-2-thio-
uracil, 5-
Methylaminomethy1-2-seleno-uracil, 5-(iso-pentenylaminomethyl)-uracil, 5-(iso-
Pentenylaminomethyl)-2-
thio-uracil, or 5-(iso-Pentenylaminomethyl)-uracil.
In other embodiments, at least one base is 5-Trideuteromethy1-6-deutero-
uracil, 5-(2-Chloro-
pheny1)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-cytosine, 5-(2-FuranyI)-
uracil, N4-methyl-cytosine, 8-
Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine,
3-Deaza-3-bromo-
adenine, or 3-Deaza-3-iodo-adenine.
- 4 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In certain embodiments, at least one base is 1-Hydroxymethyl-pseudo-uracil, 1-
(2-
Hydroxyethyl)-pseudo-uracil, 1-Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-
pseudo-uracil, 1-(2,2-
Diethoxyethyl)-pseudo-uracil, 1-(2-HydroxypropyI)-pseudo-uracil, (2R)-1-(2-
HydroxypropyI)-pseudo-uracil,
(2S)-1-(2-HydroxypropyI)-pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-
Morpholinomethyl-pseudo-
uracil, 1-Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil,
PentafluoropropyI)-pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-
Methanesulfonylmethyl-pseudo-
uracil, 1-Vinyl-pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-
uracil, 1-Propargyl-pseudo-uracil,
1-(4-FluorobenzyI)-pseudo-uracil, 1-(4-ChlorobenzyI)-pseudo-uracil, 1-(4-
BromobenzyI)-pseudo-uracil, 1-
(4-lodobenzy1)-pseudo-uracil, 1-(4-MethylbenzyI)-pseudo-uracil, 1-(4-
TrifluoromethylbenzyI)-pseudo-
uracil, 1-(4-MethoxybenzyI)-pseudo-uracil, 1-(4-TrifluoromethoxybenzyI)-pseudo-
uracil, 1-(4-
Thiomethoxybenzy1)-pseudo-uracil, 1-(4-MethanesulfonylbenzyI)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
TrimethylbenzyI)-pseudo-
uracil, 1-(4-NitrobenzyI)-pseudo-uracil, 1-(4-AzidobenzyI)-pseudo-uracil, 1-
(3,4-DimethoxybenzyI)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyI)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyI)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 143-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxyyethoxy
)-ethoxy}]propionic acid, 1-{342-(2-Aminoethoxy)-ethoxyypropionyl } pseudo-
uracil, 143-(2-{242-(2-
Aminoethoxy)-ethoxyyethoxy}-ethoxy)-propionylypseudo-uracil, 1-Biotinyl-pseudo-
uracil, or 1-Biotinyl-
PEG2-pseudo-uracil.
In some embodiments, at least one base is 5-cyclopropyl-cytosine, 5-methyl-N6-
acetyl-
cytosine, 5-(carboxymethyl)-N6-trifluoroacetyl-cytosine trifluoromethyl ester,
N6-propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-
propionyI)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-pseudo-isocytosine, or 1-hydroxy-pseudo-
isocytosine.
In other embodiments, at least one base is 1-(2,2,2-trifluoroethyl)-pseudo-
uracil.
In certain embodiments, the polynucleotide includes at least one backbone
moiety of Formula
VIII-XII:
/y3 B7Y3
B _____________________________________________________ Fi) yl __ y5,
_______ Fi) yl __ v5
U JR4 / y3 U
\ y4 / =` I I \y4 R3,
___________________________________ vl -y5 R r R5
R5 Y4 / R3
I
IR- m'R m r R2 \ R6 / y2
\
y3=R __________________________________________ Y3-P __
NI/
(4 /
c1 Y3=P y / __
\ I / a I /
Y4/q
Formula VIII Formula IX Formula X,
- 5 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
7Y3
____________________________________________ Fi) yl __ y5
U
\y4
r R3
7 y2 \
4 N I \
R'Y3=P _____________________________________________________
,
Nir4
, and =
Formula XI Formula XII
wherein the dashed line represents an optional double bond;
B is a nucleobase;
each of U and U' is, independently, 0, S, N(RU)nu, or C(RU)nu, wherein nu is
an integer from 0
to 2 and each Ru is, independently, H, halo, or optionally substituted C1-C6
alkyl;
each of R1', R2', R1", R2", R1, R3', R4, R6, R6, and R7 is, independently, H,
halo, hydroxy, thiol,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6alkynyl,
optionally substituted C1-C6
heteroalkyl, optionally substituted C2-C6heteroalkenyl, optionally substituted
C2-C6heteroalkynyl,
optionally substituted amino, azido, optionally substituted C6-C10 aryl; or R6
can join together with one or
more of R1', R1", R2', or R2" to form optionally substituted C1-C6alkylene or
optionally substituted C1-C6
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted C3-C10 carbocycle or an optionally substituted C3-C9heterocycly1;
or R4 can join together with
one or more of R1', R1", R2', R2", R3, or R6to form optionally substituted C1-
C6alkylene or optionally
substituted C1-C6 heteroalkylene and, taken together with the carbons to which
they are attached, provide
an optionally substituted C3-C10 carbocycle or an optionally substituted C3-
C9heterocycly1;
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6
alkynyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C2-
C6heteroalkenyl, optionally
substituted C2-C6heteroalkynyl, optionally substituted amino, azido,
optionally substituted C6-C10 aryl; or
R3 can join together with one or more of R1', R1", R2', R2", and, taken
together with the carbons to which
they are attached, provide an optionally substituted C3-C10 carbocycle or an
optionally substituted C2-C9
heterocyclyl; wherein if said optional double bond is present, R3 is absent;
each of m' and m" is, independently, an integer from 0 to 3;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, 0, S, Se, -NR-,
optionally substituted
C1-C6alkylene, or optionally substituted C1-C6 heteroalkylene, wherein RN1 is
H, optionally substituted C1-
C6 alkyl, optionally substituted C2-C6alkenyl, optionally substituted C2-
C6alkynyl, optionally substituted C6-
C10 aryl, or absent;
each of Y4 is, independently, H, hydroxyl, protected hydroxyl, halo, thiol,
boranyl, optionally
substituted C1-C6 alkyl, optionally substituted C2-C6alkenyl, optionally
substituted C2-C6alkynyl, optionally
substituted C1-C6 heteroalkyl, optionally substituted C2-C6heteroalkenyl,
optionally substituted C2-C6
heteroalkynyl, optionally substituted amino, or absent; and
Y6 is 0, S, Se, optionally substituted C1-C6alkylene, or optionally
substituted C1-C6
heteroalkylene.
In some embodiments, the polynucleotide further includes:
- 6 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(a) a 5' UTR, e.g., comprising at least one Kozak sequence;
(b) a 3' UTR; and
(c) at least one 5' cap structure.
In other embodiments, the at least one 5' cap structure is Cap0, Cap1, ARCA,
inosine, N1-
methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-
amino-guanosine, LNA-
guanosine, or 2-azido-guanosine.
In certain embodiments, the polynucleotide further includes a poly-A tail.
In some embodiments, the polynucleotide encodes a protein of interest.
In other embodiments, the polynucleotide is purified.
In certain embodiments, the polynucleotide is codon optimized.
In another aspect, the invention features an isolated polynucleotide encoding
a polypeptide of
interest, the isolated polynucleotide including:
(a) a 5' UTR, e.g., comprising at least one Kozak sequence;
(b) a 3' UTR; and
(c) at least one 5' cap structure,
wherein at least one base is 1-Methyl-3-(3-amino-3-carboxypropyl)pseudo-
uracil, 5-Oxyacetic
acid-methyl ester-uracil, 5-Trifluoromethyl-cytosine, 5-Trifluoromethyl-
uracil, 5-
Carboxymethylaminomethy1-2-thio-uracil, 5-Methylaminomethy1-2-thio-uracil, 5-
Methoxy-carbonyl-methyl-
uracil, 5-Oxyacetic acid-uracil, 3-(3-Amino-3-carboxypropyI)-uracil, 2-Amino-
adenine, 8-Aza-adenine,
Xanthosine, 5-Bromo-cytosine, 5-Aminoallyl-cytosine, 5-iodo-cytosine, 8-bromo-
adenine, 8-bromo-
guanine, N4-Benzoyl-cytosine, N4-Amino-cytosine, N6-Bz-adenine, N2-isobutyl-
guanine, 5-
Methylaminomethy1-2-thio-uracil, 5-Carbamoylmethyl-uracil, 1-Methyl-3-(3-amino-
3-carboxypropyl)
pseudo-uracil, 5-Methyldihydro-uracil, 5-(1-propynyl)cytosine, 5-
Ethynylcytosine, 5-vinyl-uracil, (Z)-5-(2-
Bromo-vinyl)-uracil, (E)-5-(2-Bromo-vinyl)-uracil, 5-Methoxy-cytosine, 5-
Formyl-uracil, 5-Cyano-uracil, 5-
Dimethylamino-uracil, 5-Cyano-cytosine, 5-Phenylethynyl-uracil, (E)-5-(2-Bromo-
vinyl)-cytosine, 2-
Mercapto-adenine, 2-Azido-adenine, 2-Fluoro-adenine, 2-Chloro-adenine, 2-Bromo-
adenine, 2-lodo-
adenine, 7-Amino-1H-pyrazolo[4,3-d]pyrimidine, 2,4-dihydropyrazolo[4,3-
d]pyrimidin-7-one, 2,4-
dihydropyrazolo[4,3-d]pyrimidine-5,7-dione, pyrrolosine, 9-Deaza-adenine, 9-
Deaza-guanine, 3-Deaza-
adenine, 3-Deaza-3-chloro-adenine, 1-Deaza-adenine, 5-vinyl-cytosine, 5-phenyl-
cytosine, 5-
difluoromethyl-cytosine, 5-(1-propynyI)-uracil, 5-(1-propynyI)-cytosine, or 5-
methoxymethyl-cytosine.
In some embodiments, at least one base is 1-Methyl-3-(3-amino-3-
carboxypropyl)pseudo-
uracil.
In other embodiments, at least one base is 5-Oxyacetic acid-methyl ester-
uracil, 5-
Trifluoromethyl-cytosine, 5-Trifluoromethyl-uracil, 5-Carboxymethylaminomethy1-
2-thio-uracil, 5-
Methylaminomethy1-2-thio-uracil, 5-Methoxy-carbonyl-methyl-uracil, 5-Oxyacetic
acid-uracil, or 3-(3-
Amino-3-carboxypropy1)-uracil.
In certain embodiments, at least one base is 2-Amino-adenine, 8-Aza-adenine,
Xanthosine, 5-
Bromo-cytosine, or 5-Aminoallyl-cytosine.
In some embodiments, at least one base is 5-iodo-cytosine, 8-bromo-adenine, 8-
bromo-
guanine, N4-Benzoyl-cytosine, N4-Amino-cytosine, N6-Bz-adenine, or N2-isobutyl-
guanine.
- 7 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In other embodiments, at least one base is 5-Methylaminomethy1-2-thio-uracil,
5-
Carbamoylmethyl-uracil, 1-Methyl-3-(3-amino-3-carboxypropyl) pseudo-uracil, or
5-Methyldihydro-uracil.
In certain embodiments, at least one base is 5-(1-propynyl)cytosine, 5-
Ethynylcytosine, 5-
vinyl-uracil, (Z)-5-(2-Bromo-vinyl)-uracil, (E)-5-(2-Bromo-vinyl)-uracil, 5-
Methoxy-cytosine, 5-Formyl-uracil,
5-Cyano-uracil, 5-Dimethylamino-uracil, 5-Cyano-cytosine, 5-Phenylethynyl-
uracil, (E)-5-(2-Bromo-vinyl)-
cytosine, 2-Mercapto-adenine, 2-Azido-adenine, 2-Fluoro-adenine, 2-Chloro-
adenine, 2-Bromo-adenine,
2-lodo-adenine, 7-Amino-1H-pyrazolo[4,3-d]pyrimidine, 2,4-dihydropyrazolo[4,3-
d]pyrimidin-7-one, 2,4-
dihydropyrazolo[4,3-d]pyrimidine-5,7-dione, pyrrolosine, 9-Deaza-adenine, 9-
Deaza-guanine, 3-Deaza-
adenine, 3-Deaza-3-chloro-adenine, or 1-Deaza-adenine.
In some embodiments, at least one base is 5-methoxy-uracil, 5-vinyl-cytosine,
5-phenyl-
cytosine, 5-difluoromethyl-cytosine, or 5-methoxymethyl-cytosine.
In other embodiments, at least one base is 5-bromo-cytosine.
In certain embodiments, the polynucleotide further includes a poly-A tail.
In some embodiments, the polynucleotide is purified.
In other embodiments, the at least one 5' cap structure is Cap0, Cap1, ARCA,
inosine, N1-
methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-
amino-guanosine, LNA-
guanosine, or 2-azido-guanosine.
In certain embodiments, the polynucleotide is codon optimized.
In another aspect, the invention features a compound of Formula I:
A-B,
Formula I
wherein A is:
/Y3 -1,, /y3
ii ''',-:._
Y6 _____ P Y1 __ Y5 y6 __ P vl __ y5 i
U \ R4 / y3 R3' U
1\' l'r?
I '
I
's ¨ --- ' u
I I A y4 / R3 , / R4
7r R3' R1
i / R1\ y6 __________ p y 1 : y5
R1 "
R5 =R4 r R57'''
'7*.
' \ p2"/ õ \Y4 'r R3 __6 ________________________ 2"
R6 I R
7 i R2' m' ¨ m r R7 y2 \ 7 y2 )
I _________________________________________________________________ i
y3 ¨ p y6 6
Y3 ________________ Y3 P ___ y
y6
I ID
I \ I
\ Y4 /q / a , \ y4/ , xi4 q
I a
Formula ll Formula Ill Formula IV,
7Y3 22-z_
ii
y6 __________________________________________ p v 1 __ y5 /
'1 '
U \y4 /
/ r R3
HN ¨Y \ 5 7 y2 )
R'
\
L"-----:%-11 I
y3=P ______________________________________________________
I
w4 Y6
OH , or T a =
,
Formula V Formula VI
wherein the dashed line represents an optional double bond;
- 8 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of U and U' is, independently, 0, S, N(RU)nu, or C(RU)nu, wherein nu is
an integer from 0
to 2 and each Ru is, independently, H, halo, or optionally substituted C1-C6
alkyl;
each of R1', R2', R1", R1, R2", R3', R4, R6, R6, and R7 is, independently, H,
halo, hydroxy, thiol,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6alkynyl,
optionally substituted C1-C6
heteroalkyl, optionally substituted C2-C6heteroalkenyl, optionally substituted
C2-C6heteroalkynyl,
optionally substituted amino, azido, optionally substituted C6-C10 aryl; or R6
can join together with one or
more of R1', R1", R2', or R2" to form optionally substituted C1-C6alkylene or
optionally substituted C1-C6
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted C3-C10 carbocycle or an optionally substituted C3-C9heterocycly1;
or R4 can join together with
one or more of R1', R1", R2', R2", R3, or R6to form optionally substituted C1-
C6alkylene or optionally
substituted C1-C6 heteroalkylene and, taken together with the carbons to which
they are attached, provide
an optionally substituted C3-C10 carbocycle or an optionally substituted C3-
C9heterocycly1;
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6
alkynyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C2-
C6heteroalkenyl, optionally
substituted C2-C6heteroalkynyl, optionally substituted amino, azido,
optionally substituted C6-C10 aryl; or
R3 can join together with one or more of R1', R1", R2', R2", and, taken
together with the carbons to which
they are attached, provide an optionally substituted C3-C10 carbocycle or an
optionally substituted C3-C9
heterocyclyl; wherein if said optional double bond is present, R3 is absent;
each of m' and m" is, independently, an integer from 0 to 3;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, 0, S, Se, -NR-,
optionally substituted
C1-C6alkylene, or optionally substituted C1-C6 heteroalkylene, wherein RN1 is
H, optionally substituted C1-
C6 alkyl, optionally substituted C2-C6alkenyl, optionally substituted C2-
C6alkynyl, optionally substituted C6-
C10 aryl, or absent;
each of Y4 and Y6 is, independently, H, hydroxyl, protected hydroxyl, halo,
thiol, boranyl,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6alkenyl,
optionally substituted C2-C6alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-
C6heteroalkenyl, optionally substituted
C2-C6heteroalkynyl, optionally substituted amino, or absent;
Y6 is 0, S, Se, optionally substituted C1-C6alkylene, or optionally
substituted C1-C6
heteroalkylene; and
B is 1,6-Dimethyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-(1-
propyny1)-pseudo-
uracil, 1-(optionally substituted C1-C6 Alkyl)-6-(2-propyny1)-pseudo-uracil, 1-
(optionally substituted C1-C6
Alkyl)-6-allyl-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-ethynyl-
pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-6-homoallyl-pseudo-uracil, 1-(optionally substituted
C1-C6 Alkyl)-6-vinyl-pseudo-
uracil, 1-Methy1-6-(2,2,2-Trifluoroethyl)pseudo-uracil, 1-Methy1-6-(4-
morpholino)-pseudo-uracil, 1-Methyl-
6-(4-thiomorpholino)-pseudo-uracil, 1-Methy1-6-(optionally substituted
phenyl)pseudo-uracil, 1-Methy1-6-
amino-pseudo-uracil, 1-Methyl-6-azido-pseudo-uracil, 1-Methyl-6-bromo-pseudo-
uracil, 1-Methyl-6-butyl-
pseudo-uracil, 1-Methy1-6-chloro-pseudo-uracil, 1-Methy1-6-cyano-pseudo-
uracil, 1-Methy1-6-
dimethylamino-pseudo-uracil, 1-Methy1-6-ethoxy-pseudo-uracil, 1-Methy1-6-
ethylcarboxylate-pseudo-
uracil, 1-Methy1-6-ethyl-pseudo-uracil, 1-Methy1-6-fluoro-pseudo-uracil, 1-
Methy1-6-formyl-pseudo-uracil,
1-Methy1-6-hydroxyamino-pseudo-uracil, 1-Methy1-6-hydroxy-pseudo-uracil, 1-
Methy1-6-iodo-pseudo-
- 9 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
uracil, 1-Methy1-6-iso-propyl-pseudo-uracil, 1-Methy1-6-methoxy-pseudo-uracil,
1-Methy1-6-methylamino-
pseudo-uracil, 1-Methy1-6-phenyl-pseudo-uracil, 1-Methy1-6-propyl-pseudo-
uracil, 1-Methy1-6-tert-butyl-
pseudo-uracil, 1-Methy1-6-trifluoromethoxy-pseudo-uracil, 1-Methy1-6-
trifluoromethyl-pseudo-uracil, 6-
(2,2,2-Trifluoroethyl)-pseudo-uracil, 6-(4-Morpholino)-pseudo-uracil, 6-(4-
Thiomorpholino)-pseudo-uracil,
6-(optionally substituted-Phenyl)-pseudo-uracil, 6-Amino-pseudo-uracil, 6-
Azido-pseudo-uracil, 6-Bromo-
pseudo-uracil, 6-Butyl-pseudo-uracil, 6-Chloro-pseudo-uracil, 6-Cyano-pseudo-
uracil, 6-Dimethylamino-
pseudo-uracil, 6-Ethoxy-pseudo-uracil, 6-Ethylcarboxylate-pseudo-uracil, 6-
Ethyl-pseudo-uracil, 6-Fluoro-
pseudo-uracil, 6-Formyl-pseudo-uracil, 6-Hydroxyamino-pseudo-uracil, 6-Hydroxy-
pseudo-uracil, 6-lodo-
pseudo-uracil, 6-iso-Propyl-pseudo-uracil, 6-Methoxy-pseudo-uracil, 6-
Methylamino-pseudo-uracil, 6-
Methyl-pseudo-uracil, 6-Phenyl-pseudo-uracil, 6-Propyl-pseudo-uracil, 6-tert-
Butyl-pseudo-uracil, 6-
Trifluoromethoxy-pseudo-uracil, 6-Trifluoromethyl-pseudo-uracil, 1-(3-Amino-3-
carboxypropyl)pseudo-
uracil, 1-(2,2,2-Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-Trimethyl-
benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-
phenyl)pseudo-uracil, 1-(2-Amino-2-carboxyethyl)pseudo-uracil, 1-(2-Amino-
ethyl)pseudo-uracil, 1-(3-
Amino-propyl)pseudo-uracil, 1-(4-Amino-4-carboxybutyl)pseudo-uracil, 1-(4-
Amino-benzyl)pseudo-uracil,
1-(4-Amino-butyl)pseudo-uracil, 1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-
benzyl)pseudo-uracil, 1-
(4-Methoxy-phenyl)pseudo-uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-
benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil, 1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-
hexyl)pseudo-uracil, 1-
Aminomethyl-pseudo-uracil, 1-Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-
Cyclobutylmethyl-pseudo-
uracil, 1-Cyclobutyl-pseudo-uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-
Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-
pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-
pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-
uracil, 1-Hexyl-pseudo-
uracil, 1-iso-Propyl-pseudo-uracil 1-Pentyl-pseudo-uracil, 1-Phenyl-pseudo-
uracil, 1-Propyl-pseudo-uracil,
1-p-toluyl-pseudo-uracil, 1-tert-Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-
uracil, 3-(optionally
substituted C1-C6Alkyl)-pseudo-uracil, Pseudo-uracil-N1-2-ethanoic acid,
Pseudo-uracil-N1-3-propionic
acid, Pseudo-uracil-N1-4-butanoic acid, Pseudo-uracil-N1-5-pentanoic acid,
Pseudo-uracil-N1-6-hexanoic
acid, Pseudo-uracil-N1-7-heptanoic acid, Pseudo-uracil-N1-methyl-p-benzoic
acid, 6-phenyl-pseudo-
uracil, 6-azido-pseudo-uracil, Pseudo-uracil-N1-p-benzoic acid, N3-Methyl-
pseudo-uracil, 5-Methyl-
amino-methyl-uracil, 5-Carboxy-methyl-amino-methyl-uracil, 5-
(carboxyhydroxymethyl)uracil methyl ester
5-(carboxyhydroxymethyl)uracil, 2-anhydro-cytosine, 2-anhydro-uracil, 5-
Methoxycarbonylmethy1-2-thio-
uracil, 5-Methylaminomethy1-2-seleno-uracil, 5-(iso-Pentenylaminomethyl)-
uracil, 5-(iso-
Pentenylaminomethyl)- 2-thio-uracil, 5-(iso-Pentenylaminomethyl)-uracil, 5-
Trideuteromethy1-6-deutero-
uracil, 5-(2-Chloro-phenyl)-2-thio-cytosine, 5-(4-Amino-phenyl)-2-thio-
cytosine, 5-(2-Furany1)-uracil, 8-
Trifluoromethyl-adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine,
3-Deaza-3-bromo-
adenine, 3-Deaza-3-iodo-adenine, 1-Hydroxymethyl-pseudo-uracil, 1-(2-
Hydroxyethyl)-pseudo-uracil, 1-
Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-pseudo-uracil, 1-(2,2-
Diethoxyethyl)-pseudo-uracil, 1-
(2-Hydroxypropy1)-pseudo-uracil, (2R)-1-(2-Hydroxypropy1)-pseudo-uracil, (2S)-
1-(2-Hydroxypropy1)-
pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-Morpholinomethyl-pseudo-uracil,
1-
Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil, 1-
(2,2,3,3,3-Pentafluoropropy1)-
pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-Methanesulfonylmethyl-
pseudo-uracil, 1-Vinyl-
pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-uracil, 1-Propargyl-
pseudo-uracil, 1-(4-
- 10 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
FluorobenzyI)-pseudo-uracil, 1-(4-ChlorobenzyI)-pseudo-uracil, 1-(4-
BromobenzyI)-pseudo-uracil, 1-(4-
lodobenzyI)-pseudo-uracil, 1-(4-MethylbenzyI)-pseudo-uracil, 1-(4-
TrifluoromethylbenzyI)-pseudo-uracil,
1-(4-MethoxybenzyI)-pseudo-uracil, 1-(4-TrifluoromethoxybenzyI)-pseudo-uracil,
1-(4-
Thiomethoxybenzy1)-pseudo-uracil, 1-(4-MethanesulfonylbenzyI)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
TrimethylbenzyI)-pseudo-
uracil, 1-(4-NitrobenzyI)-pseudo-uracil, 1-(4-AzidobenzyI)-pseudo-uracil, 1-
(3,4-DimethoxybenzyI)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyI)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyI)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 143-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxyyethoxy
)-ethoxy}]propionic acid, 1-{342-(2-Aminoethoxy)-ethoxyypropionyl } pseudo-
uracil, 143-(2-{242-(2-
Aminoethoxy)-ethoxyyethoxy}-ethoxy)-propionylypseudo-uracil, 1-Biotinyl-pseudo-
uracil, 1-Biotinyl-
PEG2-pseudo-uracil, 5-cyclopropyl-cytosine, 5-methyl-N6-acetyl-cytosine, 5-
(carboxymethyl)-N6-
trifluoroacetyl-cytosine trifluoromethyl ester, N6-propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-
trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-propionyI)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-
pseudo-isocytosine, 1-hydroxy-pseudo-isocytosine, or 1-(2,2,2-trifluoroethyl)-
pseudo-uracil;
or a salt thereof.
In some embodiments, B is 1,6-Dimethyl-pseudo-uracil, 1-(optionally
substituted C1-C6 Alkyl)-
6-(1-propyny1)-pseudo-uracil, 1-(optionally substituted C1-C6 Alkyl)-6-(2-
propyny1)-pseudo-uracil, 1-
(optionally substituted C1-C6 Alkyl)-6-allyl-pseudo-uracil, 1-(optionally
substituted C1-C6Alkyl)-6-ethynyl-
pseudo-uracil, 1-(optionally substituted C1-C6Alkyl)-6-homoallyl-pseudo-
uracil, 1-(optionally substituted
C1-C6 Alkyl)-6-vinyl-pseudo-uracil, 1-Methy1-6-(2,2,2-Trifluoroethyl)pseudo-
uracil, 1-Methy1-6-(4-
morpholino)-pseudo-uracil, 1-Methyl-6-(4-thiomorpholino)-pseudo-uracil, 1-
Methyl-6-(optionally
substituted phenyl)pseudo-uracil, 1-Methy1-6-amino-pseudo-uracil, 1-Methy1-6-
azido-pseudo-uracil, 1-
Methy1-6-bromo-pseudo-uracil, 1-Methy1-6-butyl-pseudo-uracil, 1-Methy1-6-
chloro-pseudo-uracil, 1-
Methy1-6-cyano-pseudo-uracil, 1-Methy1-6-dimethylamino-pseudo-uracil, 1-Methy1-
6-ethoxy-pseudo-uracil,
1-Methy1-6-ethylcarboxylate-pseudo-uracil, 1-Methy1-6-ethyl-pseudo-uracil, 1-
Methy1-6-fluoro-pseudo-
uracil, 1-Methy1-6-formyl-pseudo-uracil, 1-Methy1-6-hydroxyamino-pseudo-
uracil, 1-Methy1-6-hydroxy-
pseudo-uracil, 1-Methyl-6-iodo-pseudo-uracil, 1-Methyl-6-iso-propyl-pseudo-
uracil, 1-Methy1-6-methoxy-
pseudo-uracil, 1-Methy1-6-methylamino-pseudo-uracil, 1-Methy1-6-phenyl-pseudo-
uracil, 1-Methy1-6-
propyl-pseudo-uracil, 1-Methy1-6-tert-butyl-pseudo-uracil, 1-Methy1-6-
trifluoromethoxy-pseudo-uracil, 1-
Methy1-6-trifluoromethyl-pseudo-uracil, 6-(2,2,2-Trifluoroethyl)-pseudo-
uracil, 6-(4-Morpholino)-pseudo-
uracil, 6-(4-Thiomorpholino)-pseudo-uracil, 6-(Substituted-Phenyl)-pseudo-
uracil, 6-Amino-pseudo-uracil,
6-Azido-pseudo-uracil, 6-Bromo-pseudo-uracil, 6-Butyl-pseudo-uracil, 6-Chloro-
pseudo-uracil, 6-Cyano-
pseudo-uracil, 6-Dimethylamino-pseudo-uracil, 6-Ethoxy-pseudo-uracil, 6-
Ethylcarboxylate-pseudo-uracil,
6-Ethyl-pseudo-uracil, 6-Fluoro-pseudo-uracil, 6-Formyl-pseudo-uracil, 6-
Hydroxyamino-pseudo-uracil, 6-
Hydroxy-pseudo-uracil, 6-lodo-pseudo-uracil, 6-iso-Propyl-pseudo-uracil, 6-
Methoxy-pseudo-uracil, 6-
Methylamino-pseudo-uracil, 6-Methyl-pseudo-uracil, 6-Phenyl-pseudo-uracil, 6-
Phenyl-pseudo-uracil, 6-
Propyl-pseudo-uracil, 6-tert-Butyl-pseudo-uracil, 6-Trifluoromethoxy-pseudo-
uracil, 6-Trifluoromethyl-
- 11 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
pseudo-uracil, 1-(3-Amino-3-carboxypropyl)pseudo-uracil, 1-(2,2,2-
Trifluoroethyl)-pseudo-uracil, 1-(2,4,6-
Trimethyl-benzyl)pseudo-uracil, 1-(2,4,6-Trimethyl-phenyl)pseudo-uracil, 1-(2-
Amino-2-
carboxyethyl)pseudo-uracil, 1-(2-Amino-ethyl)pseudo-uracil, 1-(3-Amino-
propyl)pseudo-uracil, 1-(4-
Amino-4-carboxybutyl)pseudo-uracil, 1-(4-Amino-benzyl)pseudo-uracil, 1-(4-
Amino-butyl)pseudo-uracil,
1-(4-Amino-phenyl)pseudo-uracil, 1-(4-Methoxy-benzyl)pseudo-uracil, 1-(4-
Methoxy-phenyl)pseudo-
uracil, 1-(4-Methyl-benzyl)pseudo-uracil, 1-(4-Nitro-benzyl)pseudo-uracil, 1(4-
Nitro-phenyl)pseudo-uracil,
1-(5-Amino-pentyl)pseudo-uracil, 1-(6-Amino-hexyl)pseudo-uracil, 1-Aminomethyl-
pseudo-uracil, 1-
Benzyl-pseudo-uracil, 1-Butyl-pseudo-uracil, 1-Cyclobutylmethyl-pseudo-uracil,
1-Cyclobutyl-pseudo-
uracil, 1-Cycloheptylmethyl-pseudo-uracil, 1-Cycloheptyl-pseudo-uracil, 1-
Cyclohexylmethyl-pseudo-
uracil, 1-Cyclohexyl-pseudo-uracil, 1-Cyclooctylmethyl-pseudo-uracil, 1-
Cyclooctyl-pseudo-uracil, 1-
Cyclopentylmethyl-pseudo-uracil, 1-Cyclopentyl-pseudo-uracil, 1-
Cyclopropylmethyl-pseudo-uracil, 1-
Cyclopropyl-pseudo-uracil, 1-Ethyl-pseudo-uracil, 1-Hexyl-pseudo-uracil, 1-iso-
Propyl-pseudo-uracil, 1-
Pentyl-pseudo-uracil, 1-Phenyl-pseudo-uracil, 1-Propyl-pseudo-uracil, 1-p-
tolyl-pseudo-uracil, 1-tert-
Butyl-pseudo-uracil, 1-Trifluoromethyl-pseudo-uracil, 3-(optionally
substituted C1-C6 Alkyl)-pseudo-uracil,
Pseudo-uracil-N1-2-ethanoic acid, Pseudo-uracil-N1-3-propionic acid, Pseudo-
uracil-N1-4-butanoic acid,
Pseudo-uracil-N1-5-pentanoic acid, Pseudo-uracil-N1-6-hexanoic acid, Pseudo-
uracil-N1-7-heptanoic
acid, Pseudo-uracil-N1-methyl-p-benzoic acid, 6-phenyl-pseudo-uracil, 6-azido-
pseudo-uracil, or Pseudo-
uracil-N1-p-benzoic acid.
In other embodiments, B is N3-Methyl-pseudo-uracil, 5-Methyl-amino-methyl-
uracil, 5-
Carboxy-methyl-amino-methyl-uracil, 5-(carboxyhydroxymethyl)uracil methyl
ester, or 5-
(carboxyhydroxymethyl)uracil.
In certain embodiments, B is 2-anhydro-cytosine or 2-anhydro-uracil.
In some embodiments, B is 5-Methoxycarbonylmethy1-2-thio-uracil, 5-
Methylaminomethy1-2-
seleno-uracil, 5-(iso-Pentenylaminomethyl)-uracil, 5-(iso-Pentenylaminomethyl)-
2-thio-uracil, or 5-(iso-
Pentenylaminomethyl)-uracil.
In other embodiments, B is 5-Trideuteromethy1-6-deutero-uracil, 5-(2-Chloro-
pheny1)-2-thio-
cytosine, 5-(4-Amino-phenyl)-2-thio-cytosine, 5-(2-FuranyI)-uracil, N4-methyl-
cytosine, 8-Trifluoromethyl-
adenine, 2-Trifluoromethyl-adenine, 3-Deaza-3-fluoro-adenine, 3-Deaza-3-bromo-
adenine, or 3-Deaza-3-
iodo-adenine.
In certain embodiments, B is 1-Hydroxymethyl-pseudo-uracil, 1-(2-Hydroxyethyl)-
pseudo-
uracil, 1-Methoxymethyl-pseudo-uracil, 1-(2-Methoxyethyl)-pseudo-uracil, 1-
(2,2-Diethoxyethyl)-pseudo-
uracil, 1-(2-HydroxypropyI)-pseudo-uracil, (2R)-1-(2-HydroxypropyI)-pseudo-
uracil, (2S)-1-(2-
Hydroxypropy1)-pseudo-uracil, 1-Cyanomethyl-pseudo-uracil, 1-Morpholinomethyl-
pseudo-uracil, 1-
Thiomorpholinomethyl-pseudo-uracil, 1-Benzyloxymethyl-pseudo-uracil, 1-
(2,2,3,3,3-PentafluoropropyI)-
pseudo-uracil, 1-Thiomethoxymethyl-pseudo-uracil, 1-Methanesulfonylmethyl-
pseudo-uracil, 1-Vinyl-
pseudo-uracil, 1-Allyl-pseudo-uracil, 1-Homoallyl-pseudo-uracil, 1-Propargyl-
pseudo-uracil, 1-(4-
Fluorobenzy1)-pseudo-uracil, 1-(4-ChlorobenzyI)-pseudo-uracil, 1-(4-
BromobenzyI)-pseudo-uracil, 1-(4-
lodobenzyI)-pseudo-uracil, 1-(4-MethylbenzyI)-pseudo-uracil, 1-(4-
TrifluoromethylbenzyI)-pseudo-uracil,
1-(4-MethoxybenzyI)-pseudo-uracil, 1-(4-TrifluoromethoxybenzyI)-pseudo-uracil,
1-(4-
ThiomethoxybenzyI)-pseudo-uracil, 1-(4-MethanesulfonylbenzyI)-pseudo-uracil,
Pseudo-uracil 1-(4-
methylbenzoic acid), Pseudo-uracil 1-(4-methylbenzenesulfonic acid), 1-(2,4,6-
TrimethylbenzyI)-pseudo-
- 12-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
uracil, 1-(4-NitrobenzyI)-pseudo-uracil, 1-(4-AzidobenzyI)-pseudo-uracil, 1-
(3,4-DimethoxybenzyI)-
pseudo-uracil, 1-(3,4-Bis-trifluoromethoxybenzyI)-pseudo-uracil, 1-Acetyl-
pseudo-uracil, 1-Trifluoroacetyl-
pseudo-uracil, 1-Benzoyl-pseudo-uracil, 1-Pivaloyl-pseudo-uracil, 1-(3-
Cyclopropyl-prop-2-ynyI)-pseudo-
uracil, Pseudo-uracil 1-methylphosphonic acid diethyl ester, Pseudo-uracil 1-
methylphosphonic acid,
Pseudo-uracil 1-[3-(2-ethoxy)]propionic acid, Pseudo-uracil 143-{2-(2-ethoxy)-
ethoxy}] propionic acid,
Pseudo-uracil 1-[3-{2-(2-[2-ethoxy ]-ethoxy)-ethoxy}]propionic acid, Pseudo-
uracil 1-[3-{2-(2-[2-(2-ethoxy
)-ethoxy]-ethoxy )-ethoxy}]propionic acid, Pseudo-uracil 1-[3-{2-(2-[2-{2(2-
ethoxy )-ethoxy}-ethoxyyethoxy
)-ethoxy}]propionic acid, 1-{342-(2-Aminoethoxy)-ethoxyypropionyl } pseudo-
uracil, 143-(2-{242-(2-
Aminoethoxy)-ethoxyyethoxy}-ethoxy)-propionylypseudo-uracil, 1-Biotinyl-pseudo-
uracil, or 1-Biotinyl-
PEG2-pseudo-uracil.
In some embodiments, B is 5-cyclopropyl-cytosine, 5-methyl-N6-acetyl-cytosine,
5-
(carboxymethyl)-N6-trifluoroacetyl-cytosine trifluoromethyl ester, N6-
propionyl-cytosine, 5-
monofluoromethyl-cytosine, 5-trifluoromethoxy-cytosine, N6-(1,1,1-trifluoro-
propionyI)-cytosine, 4-acetyl-
pseudo-isocytosine, 1-ethyl-pseudo-isocytosine, or 1-hydroxy-pseudo-
isocytosine.
In other embodiments, B is 1-(2,2,2-trifluoroethyl)-pseudo-uracil.
In certain embodiments, A has the structure of Formula II.
In some embodiments, m' is 0.
In other embodiments, m" is 1.
In certain embodiments, R4 is hydrogen.
In some embodiments, A is:
(y3 \
y6 ________________________________ :)_y1 __ y5
1 .-rxrir,
U/,\H
y4 / \, =s`' u
/I" R3 ___________________________________________ R1
R5
--..-
7 y2
1
Y3:P _____________________________________________ y6
\
a .
Formula VII
wherein U is 0, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 1 to 2
and each Ru is,
independently, H, halo, or optionally substituted C1-C6 alkyl;
each of R1, R2, and R5 is, independently, H, halo, hydroxy, thiol, optionally
substituted C1-C6
alkyl, optionally substituted C2-C6alkynyl, optionally substituted C1-C6
heteroalkyl, optionally substituted
C2-C6 heteroalkenyl, optionally substituted C2-C6 heteroalkynyl, optionally
substituted amino, azido,
optionally substituted C6-C10 aryl; or R5 can join together with one of R1 or
R2 to form optionally
substituted C1-C6alkylene or optionally substituted C1-C6 heteroalkylene and,
taken together with the
carbons to which they are attached, provide an optionally substituted C3-C10
carbocycle or an optionally
substituted C3-C9heterocycly1; or;
-13-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
R3 is H, halo, hydroxy, thiol, optionally substituted C1-C6 alkyl, optionally
substituted C2-C6
alkynyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C2-
C6 heteroalkenyl, optionally
substituted C2-C6 heteroalkynyl, optionally substituted amino, azido,
optionally substituted C6-C10 aryl; or
R3 can join together with one of R1 or R2, and, taken together with the
carbons to which they are
attached, provide an optionally substituted C3-C10 carbocycle or an optionally
substituted C3-C9
heterocyclyl;
each of q and r is independently, an integer from 0 to 5;
each of Y1, Y2, and Y3, is, independently, hydrogen, 0, S, Se, -NR-,
optionally substituted
C1-C6alkylene, or optionally substituted C1-C6 heteroalkylene, wherein RN1 is
H, optionally substituted C1-
C6 alkyl, optionally substituted C2-C6alkenyl, optionally substituted C2-C6
alkynyl, optionally substituted C6-
C10 aryl, or absent; and
each of Y4 and Y6 is, independently, H, hydroxyl, protected hydroxyl, halo,
thiol, boranyl,
optionally substituted C1-C6 alkyl, optionally substituted C2-C6alkenyl,
optionally substituted C2-C6 alkynyl,
optionally substituted C1-C6 heteroalkyl, optionally substituted C2-C6
heteroalkenyl, optionally substituted
C2-C6heteroalkynyl, optionally substituted amino, or absent; and
Y5 is 0, S, Se, optionally substituted C1-C6alkylene, or optionally
substituted C1-C6
heteroalkylene.
In some embodiments, R2 is hydroxyl.
In other embodiments, R1 is hydrogen.
In certain embodiments, R3 is hydrogen, and R5 is hydrogen.
In some embodiments, R3 is hydrogen, and R5 is optionally substituted C2-C6
alkynyl.
In other embodiments, the optionally substituted C2-C6alkynyl is ethynyl.
In certain embodiments, R5 is hydrogen.
In some embodiments, R3 is azido or optionally substituted C2-C6 alkynyl.
In other embodiments, R3 is azido.
In certain embodiments, R3 is optionally substituted C2-C6 alkynyl, wherein
said optionally
substituted C2-C6 alkynyl is ethynyl.
In some embodiments, R3 is hydrogen and R5 is hydrogen.
In other embodiments, R1 is optionally substituted C2-C6 alkyl or optionally
substituted C2-C6
alkynyl.
In certain embodiments, R1 is optionally substituted C2-C6 alkyl, wherein said
optionally
substituted C2-C6 alkyl is trifluoromethyl.
In some embodiments, R1 is optionally substituted C2-C6 alkynyl, wherein said
optionally
substituted C2-C6 alkynyl is ethynyl.
In other embodiments, R2 is hydrogen.
In certain embodiments, R3 is hydrogen.
In some embodiments, R5 is hydrogen.
In other embodiments, R1 is halo, thiol, optionally substituted C1-C6
heteroalkyl, azido, or
amino.
In certain embodiments, halo is fluoro, chloro, bromo, or iodo.
In some embodiments, optionally substituted C1-C6 heteroalkyl is thiomethoxy.
- 14-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In other embodiments, R3 is hydrogen.
In certain embodiments, R5 is hydrogen.
In some embodiments, R1" is hydroxy.
In other embodiments, R2" is hydrogen, optionally substituted C1-C6 alkyl, or
optionally
substituted C2-C6alkynyl.
In certain embodiments, optionally substituted C1-C6 alkyl is trifluoromethyl.
In some embodiments, optionally substituted C2-C6alkynyl is ethynyl.
In other embodiments, R1" is hydrogen.
In certain embodiments, R2" is thiol, optionally substituted C1-C6heteroalkyl,
azido, or amino.
In some embodiments, optionally substituted C1-C6heteroalkyl is thiomethoxy.
In other embodiments, R1" is halo.
In certain embodiments, halo is fluoro.
In some embodiments, R2" is halo.
In other embodiments, halo is fluoro.
In certain embodiments, U is C(RU)nu.
In some embodiments, nu is 2.
In other embodiments, each Ru is hydrogen.
In certain embodiments, q is 0; and Y6 is hydroxyl.
In some embodiments, R5 is hydroxyl.
In other embodiments, Y5 is optionally substituted C1-C6alkylene.
In certain embodiments, optionally substituted C1-C6alkylene is methylene.
In some embodiments, r is 0, and Y6 is hydroxyl.
In other embodiments, r is 3; each Y1, Y3, and Y4 is OH; and Y6 is hydroxyl.
In certain embodiments, r is 3, each Y1 and Y4 is OH; and Y6 is hydroxyl.
In some embodiments, at least one Y3 is S.
In some embodiments, the nucleobase is selected from a naturally occurring
nucleobase
or a non-naturally occurring nucleobase.
In some embodiments, the naturally occurring nucleobase is selected from the
group
consisting of pseudouracil or N1-methylpseudouracil.
In some embodiments, the nucleoside is not pseudouridine (t.p) or 5-methyl-
cytidine
(m5C).
The present invention provides polynucleotides which may be isolated and/or
purified.
These polynucleotides may encode one or more polypeptides of interest and
comprise a sequence of
n number of linked nucleosides or nucleotides comprising at least one modified
nucleoside or
nucleotide as compared to the chemical structure of an A, G, U or C nucleoside
or nucleotide. The
polynucleotides may also contain a 5' UTR, e.g., comprising at least one Kozak
sequence, a 3' UTR,
and at least one 5' cap structure. The isolated polynucleotides may further
contain a poly-A tail and
may be purified.
In some embodiments, multiple modifications are included in the modified
nucleic acid or
in one or more individual nucleoside or nucleotide. For example, modifications
to a nucleoside may
include one or more modifications to the nucleobase, the sugar, and/or the
internucleoside linkage.
-15-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: pseudouridine-
alpha-thio-MP, 1-methyl-pseudouridine-alpha-thio-MP, 1-ethyl-pseudouridine-MP,
1-propyl-
pseudouridine-MP, 1-(2,2,2-trifluoroethyl)-pseudouridine-MP, 2-amino-adenine-
MP, xanthosine-MP,
5-bromo-cytidine-MP, 5-aminoallyl-cytidine-MP, or 2-aminopurine-riboside-MP.
In certain embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: pseudouridine-
alpha-thio-MP, 1-methyl-pseudouridine-alpha-thio-MP, 1-ethyl-pseudouridine-MP,
1-propyl-
pseudouridine-MP, 5-bromo-cytidine-MP, 5-aminoallyl-cytidine-MP, or 2-
aminopurine-riboside-MP.
In other embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: pseudouridine-
alpha-thio-MP, 1-methyl-pseudouridine-alpha-thio-MP, or 5-bromo-cytidine-MP.
In other embodiments, the isolated polynucleotide includes at least two
modified
nucleosides or nucleotides.
In certain embodiments having at least two modifications, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of at least one of
each of 5-bromo-cytidine-MP and 1-methyl-pseudouridine-MP.
In other embodiments having at least two modifications, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of at least one of
each of 5-bromo-cytidine-MP and pseudouridine-MP.
In some embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: 2-thio-
pseudouridine-MP, 5-trifluoromethyl-uridine-MP, 5-trifluoromethyl-cytidine-MP,
3-methyl-
pseudouridine-MP, 5-methyl-2-thio-uridine-MP, N4-methyl-cytidine-MP, 5-
hydroxymethyl-cytidine-MP,
3-methyl-cytidine-MP, 5-oxyacetic acid methyl ester-uridine-MP, 5-
methoxycarbonylmethyl-uridine-
MP, 5-methylaminomethyl-uridine-MP, 5-methoxy-uridine-MP, N1-methyl-guanosine-
MP, 8-aza-
adenine-MP, 2-thio-uridine-MP, 5-bromo-uridine-MP, 2-thio-cytidine-MP, alpha-
thio-cytidine-MP, 5-
aminoallyl-uridine-MP, alpha-thio-uridine-MP, or 4-thio-uridine-MP.
In other embodiments having at least two modifications, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of at least one of
each of 5-trifluoromethyl-cytidine-MP and 1-methyl-pseudouridine-MP; 5-
hydroxymethyl-cytidine-MP
and 1-methyl-pseudouridine-MP; 5-trifluoromethyl-cytidine-MP and pseudouridine-
MP; or N4-acetyl-
cytidine-MP and 5-methoxy-uridine-MP.
In some embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: 2-thio-
pseudouridine-MP, 5-trifluoromethyl-cytidine-MP, 5-methyl-2-thio-uridine-MP, 5-
hydroxymethyl-
cytidine-MP, 5-oxyacetic acid methyl ester-uridine-MP, 5-methoxy-uridine-MP,
N4-acetyl-cytidine-MP,
2-thio-uridine-MP, 5-bromo-uridine-MP, alpha-thio-cytidine-MP, 5-aminoallyl-
uridine-MP, or alpha-
thio-uridine-MP.
In other embodiments having at least two modifications, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of at least one of
- 16-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of 5-trifluoromethyl-cytidine-MP and 1-methyl-pseudouridine-MP or 5-
hydroxymethyl-cytidine-
MP and 1-methyl-pseudouridine-MP.
In some embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: 2-thio-
pseudouridine-MP, 5-trifluoromethyl-cytidine-MP, 5-methyl-2-thio-uridine-MP,
N4-methyl-cytidine-MP,
5-hydroxymethyl-cytidine-MP, 5-oxyacetic acid methyl ester-uridine-MP, 5-
methoxycarbonylmethyl-
uridine-MP, 5-methoxy-uridine-MP, 2-thio-uridine-MP, 5-bromo-uridine-MP, alpha-
thio-cytidine-MP, 5-
aminoallyl-uridine-MP, or alpha-thio-uridine-MP.
In some embodiments having at least one modification, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of: 2-thio-
pseudouridine-MP, 5-trifluoromethyl-cytidine-MP, 5-hydroxymethyl-cytidine-MP,
or 5-methoxy-uridine-
MP.
In other embodiments having at least two modifications, the polynucleotide
includes a
backbone moiety containing the nucleobase, sugar, and internucleoside linkage
of at least one of
each of N4-acetyl-cytidine-MP and 5-methoxy-uridine-MP.
The present invention also provides for pharmaceutical compositions comprising
the
modified polynucleotides described herein. These may also further include one
or more
pharmaceutically acceptable excipients selected from a solvent, aqueous
solvent, non-aqueous
solvent, dispersion media, diluent, dispersion, suspension aid, surface active
agent, isotonic agent,
thickening or emulsifying agent, preservative, lipid, lipidoids liposome,
lipid nanoparticle, core-shell
nanoparticles, polymer, lipoplexed peptide, protein, cell, hyaluronidase, and
mixtures thereof.
Methods of using the polynucleotides and modified nucleic acids of the
invention are also
provided. In this instance, the polynucleotides may be formulated by any means
known in the art or
administered via any of several routes including injection by intradermal,
subcutaneous or
intramuscular means.
Administration of the modified nucleic acids of the invention may be via two
or more equal
or unequal split doses. In some embodiments, the level of the polypeptide
produced by the subject by
administering split doses of the polynucleotide is greater than the levels
produced by administering
the same total daily dose of polynucleotide as a single administration.
Detection of the modified nucleic acids or the encoded polypeptides may be
performed in
the bodily fluid of the subject or patient where the bodily fluid is selected
from the group consisting of
peripheral blood, serum, plasma, ascites, urine, cerebrospinal fluid (CSF),
sputum, saliva, bone
marrow, synovial fluid, aqueous humor, amniotic fluid, cerumen, breast milk,
broncheoalveolar lavage
fluid, semen, prostatic fluid, cowper's fluid or pre-ejaculatory fluid, sweat,
fecal matter, hair, tears, cyst
fluid, pleural and peritoneal fluid, pericardial fluid, lymph, chyme, chyle,
bile, interstitial fluid, menses,
pus, sebum, vomit, vaginal secretions, mucosal secretion, stool water,
pancreatic juice, lavage fluids
from sinus cavities, bronchopulmonary aspirates, blastocyl cavity fluid, and
umbilical cord blood.
In some embodiments, administration is according to a dosing regimen which
occurs over
the course of hours, days, weeks, months, or years and may be achieved by
using one or more
devices selected from multi-needle injection systems, catheter or lumen
systems, and ultrasound,
electrical or radiation based systems.
-17-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
The names of nucleobases correspond to the name given to the base when part of
a
nucleoside or nucleotide. For example, "pseudo-uracil" refers to the
nucleobase of pseudouridine
and "pseudo-isocytosine" refers to the nucleobase of pseudoisocytidine.
Unless otherwise defined, all technical and scientific terms used herein have
the same
meaning as commonly understood by one of ordinary skill in the art to which
this invention belongs.
Methods and materials are described herein for use in the present disclosure;
other, suitable methods
and materials known in the art can also be used. The materials, methods, and
examples are
illustrative only and not intended to be limiting. All publications, patent
applications, patents,
sequences, database entries, and other references mentioned herein are
incorporated by reference
in their entirety. In case of conflict, the present specification, including
definitions, will control.
Other features and advantages of the present disclosure will be apparent from
the
following detailed description and figures, and from the claims.
DETAILED DESCRIPTION
The present disclosure provides, inter alia, modified nucleosides, modified
nucleotides,
and modified nucleic acids that exhibit improved therapeutic properties
including, but not limited to, a
reduced innate immune response when introduced into a population of cells.
As there remains a need in the art for therapeutic modalities to address the
myriad of
barriers surrounding the efficacious modulation of intracellular translation
and processing of nucleic
acids encoding polypeptides or fragments thereof, the inventors have shown
that certain modified
mRNA sequences have the potential as therapeutics with benefits beyond just
evading, avoiding or
diminishing the immune response.
The present invention addresses this need by providing nucleic acid based
compounds or
polynucleotides which encode a polypeptide of interest (e.g., modified mRNA)
and which have
structural and/or chemical features that avoid one or more of the problems in
the art, for example,
features which are useful for optimizing nucleic acid-based therapeutics while
retaining structural and
functional integrity, overcoming the threshold of expression, improving
expression rates, half life
and/or protein concentrations, optimizing protein localization, and avoiding
deleterious bio-responses
such as the immune response and/or degradation pathways.
Polypeptides of interest, according to the present invention, may be selected
from any of
those disclosed in US 2013/0259924, US 2013/0259923, WO 2013/151663, WO
2013/151669, WO
2013/151670, WO 2013/151664, WO 2013/151665, WO 2013/151736, U.S. Provisional
Patent
Application No 61/618,862, U.S. Provisional Patent Application No 61/681,645,
U.S. Provisional
Patent Application No 61/618,873, U.S. Provisional Patent Application No
61/681,650, U.S.
Provisional Patent Application No 61/618,878, U.S. Provisional Patent
Application No 61/681,654,
U.S. Provisional Patent Application No 61/618,885, U.S. Provisional Patent
Application No
61/681,658, U.S. Provisional Patent Application No 61/618,911 s, U.S.
Provisional Patent Application
No 61/681,667, U.S. Provisional Patent Application No 61/618,922, U.S.
Provisional Patent
Application No 61/681,675, U.S. Provisional Patent Application No 61/618,935,
U.S. Provisional
Patent Application No 61/681,687, U.S. Provisional Patent Application No
61/618,945, U.S.
Provisional Patent Application No 61/681,696, U.S. Provisional Patent
Application No 61/618,953,and
-18-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
U.S. Provisional Patent Application No 61/681,704, the polypeptides of which
are incorporated herein
by reference in their entirety.
Provided herein, in part, are polynucleotides encoding polypeptides of
interest which have
been chemically modified to improve one or more of the stability and/or
clearance in tissues, receptor
uptake and/or kinetics, cellular access by the compositions, engagement with
translational machinery,
mRNA half-life, translation efficiency, immune evasion, protein production
capacity, secretion
efficiency (when applicable), accessibility to circulation, protein half-life
and/or modulation of a cell's
status, function and/or activity.
The modified nucleosides, nucleotides and nucleic acids of the invention,
including the
combination of modifications taught herein have superior properties making
them more suitable as
therapeutic modalities.
It has been determined that the "all or none" model in the art is sorely
insufficient to
describe the biological phenomena associated with the therapeutic utility of
modified mRNA. The
present inventors have determined that to improve protein production, one may
consider the nature of
the modification, or combination of modifications, the percent modification
and survey more than one
cytokine or metric to determine the efficacy and risk profile of a particular
modified mRNA.
In one aspect of the invention, methods of determining the effectiveness of a
modified
mRNA as compared to unmodified involves the measure and analysis of one or
more cytokines
whose expression is triggered by the administration of the exogenous nucleic
acid of the invention.
These values are compared to administration of an unmodified nucleic acid or
to a standard metric
such as cytokine response, PolyIC, R-848 or other standard known in the art.
One example of a standard metric developed herein is the measure of the ratio
of the level
or amount of encoded polypeptide (protein) produced in the cell, tissue or
organism to the level or
amount of one or more (or a panel) of cytokines whose expression is triggered
in the cell, tissue or
organism as a result of administration or contact with the modified nucleic
acid. Such ratios are
referred to herein as the Protein:Cytokine Ratio or "PC" Ratio. The higher the
PC ratio, the more
efficacious the modified nucleic acid (polynucleotide encoding the protein
measured). Preferred PC
Ratios, by cytokine, of the present invention may be greater than 1, greater
than 10, greater than 100,
greater than 1000, greater than 10,000 or more. Modified nucleic acids having
higher PC Ratios than
a modified nucleic acid of a different or unmodified construct are preferred.
The PC ratio may be further qualified by the percent modification present in
the
polynucleotide. For example, normalized to a 100% modified nucleic acid, the
protein production as a
function of cytokine (or risk) or cytokine profile can be determined.
In one embodiment, the present invention provides a method for determining,
across
chemistries, cytokines or percent modification, the relative efficacy of any
particular modified
polynucleotide by comparing the PC Ratio of the modified nucleic acid
(polynucleotide).
In another embodiment, the chemically modified mRNA are substantially non
toxic and
non mutagenic.
In one embodiment, the modified nucleosides, modified nucleotides, and
modified nucleic
acids can be chemically modified, thereby disrupting interactions, which may
cause innate immune
responses. Further, these modified nucleosides, modified nucleotides, and
modified nucleic acids can
-19-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
be used to deliver a payload, e.g., detectable or therapeutic agent, to a
biological target. For
example, the nucleic acids can be covalently linked to a payload, e.g. a
detectable or therapeutic
agent, through a linker attached to the nucleobase or the sugar moiety. The
compositions and
methods described herein can be used, in vivo and in vitro, both
extracellularly and intracellularly, as
well as in assays such as cell free assays.
In another aspect, the present disclosure provides chemical modifications
located on the
sugar moiety of the nucleotide.
In another aspect, the present disclosure provides chemical modifications
located on the
phosphate backbone of the nucleic acid.
In another aspect, the present disclosure provides nucleotides that contain
chemical
modifications, wherein the nucleotide reduces the cellular innate immune
response, as compared to
the cellular innate immune induced by a corresponding unmodified nucleic acid.
In another aspect, the present disclosure provides compositions comprising a
compound
as described herein. In some embodiments, the composition is a reaction
mixture. In some
embodiments, the composition is a pharmaceutical composition. In some
embodiments, the
composition is a cell culture. In some embodiments, the composition further
comprises an RNA
polymerase and a cDNA template. In some embodiments, the composition further
comprises a
nucleotide selected from the group consisting of adenosine, cytosine,
guanosine, and uracil.
In a further aspect, the present disclosure provides methods of making a
pharmaceutical
formulation comprising a physiologically active secreted protein, comprising
transfecting a first
population of human cells with the pharmaceutical nucleic acid made by the
methods described
herein, wherein the secreted protein is active upon a second population of
human cells.
In some embodiments, the secreted protein is capable of interacting with a
receptor on the
surface of at least one cell present in the second population.
In certain embodiments, provided herein are combination therapeutics
containing one or
more modified nucleic acids containing translatable regions that encode for a
protein or proteins that
boost a mammalian subject's immunity along with a protein that induces
antibody-dependent cellular
toxicity.
In one embodiment, it is intended that the compounds of the present disclosure
are stable.
It is further appreciated that certain features of the present disclosure,
which are, for clarity, described
in the context of separate embodiments, can also be provided in combination in
a single embodiment.
Conversely, various features of the present disclosure which are, for brevity,
described in the context
of a single embodiment, can also be provided separately or in any suitable
subcombination.
Modified Nucleotides, Nucleosides and Polynucleotides of the invention
Herein, in a nucleotide, nucleoside or polynucleotide (such as the nucleic
acids of the
invention, e.g., mRNA molecule), the terms "modification" or, as appropriate,
"modified" refer to
modification with respect to A, G, U or C ribonucleotides. Generally, herein,
these terms are not
intended to refer to the ribonucleotide modifications in naturally occurring
5'-terminal mRNA cap
moieties. In a polypeptide, the term "modification" refers to a modification
as compared to the
canonical set of 20 amino acids, moiety).
- 20 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
The modifications may be various distinct modifications. In some embodiments,
where the
nucleic acid is an mRNA, the coding region, the flanking regions and/or the
terminal regions may
contain one, two, or more (optionally different) nucleoside or nucleotide
modifications. In some
embodiments, a modified polynucleotide introduced to a cell may exhibit
reduced degradation in the
cell, as compared to an unmodified polynucleotide.
The polynucleotides can include any useful modification, such as to the sugar,
the
nucleobase, or the internucleoside linkage (e.g. to a linking phosphate / to a
phosphodiester linkage /
to the phosphodiester backbone). In certain embodiments, modifications (e.g.,
one or more
modifications) are present in each of the sugar and the internucleoside
linkage. Modifications
according to the present invention may be modifications of ribonucleic acids
(RNAs) to
deoxyribonucleic acids (DNAs), e.g., the substitution of the 2'0H of the
ribofuranosyl ring to 2'H,
threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic
acids (PNAs), locked
nucleic acids (LNAs) or hybrids thereof). Additional modifications are
described herein.
As described herein, the polynucleotides of the invention do not substantially
induce an
innate immune response of a cell into which the polynucleotide (e.g., mRNA) is
introduced. Features
of an induced innate immune response include 1) increased expression of pro-
inflammatory
cytokines, 2) activation of intracellular PRRs (RIG-I, MDA5, etc, and/or 3)
termination or reduction in
protein translation.
In certain embodiments, it may desirable for a modified nucleic acid molecule
introduced
into the cell to be degraded intracellularly. For example, degradation of a
modified nucleic acid
molecule may be preferable if precise timing of protein production is desired.
Thus, in some
embodiments, the invention provides a modified nucleic acid molecule
containing a degradation
domain, which is capable of being acted on in a directed manner within a cell.
The polynucleotides can optionally include other agents (e.g., RNAi-inducing
agents, RNAi
agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA,
tRNA, RNAs that
induce triple helix formation, aptamers, vectors, etc.). In some embodiments,
the polynucleotides
may include one or more messenger RNAs (mRNAs) having one or more modified
nucleoside or
nucleotides (i.e., modified mRNA molecules). Details for these polynucleotides
follow.
Polynucleotides
According to Aduri et al (Aduri, R. et al., AMBER force field parameters for
the naturally
occurring modified nucleotides in RNA. Journal of Chemical Theory and
Computation. 2006.
3(4):1464-75) there are 107 naturally occurring nucleosides, including 1-
methyladenosine, 2-
methylthio-N6-hydroxynorvaly1 carbamoyladenosine, 2-methyladenosine, 2-0-
ribosylphosphate
adenosine, N6-methyl-N6-threonylcarbamoyladenosine, N6-acetyladenosine, N6-
glycinylcarbamoyladenosine, N6-isopentenyladenosine, N6-methyladenosine, N6-
threonylcarbamoyladenosine, N6,N6-dimethyladenosine, N6-(cis-
hydroxyisopentenyl)adenosine, N6-
hydroxynorvalylcarbamoyladenosine, 1,2-0-dimethyladenosine, N6,2-0-
dimethyladenosine, 2-0-
methyladenosine, N6,N6,0-2-trimethyladenosine, 2-methylthio-N6-(cis-
hydroxyisopentenyl)
adenosine, 2-methylthio-N6-methyladenosine, 2-methylthio-N6-
isopentenyladenosine, 2-methylthio-
N6-threonyl carbamoyladenosine, 2-thiocytidine, 3-methylcytidine, N4-
acetylcytidine, 5-formylcytidine,
N4-methylcytidine, 5-methylcytidine, 5-hydroxymethylcytidine, lysidine, N4-
acetyl-2-0-methylcytidine,
- 21 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
5-formy1-2-0-methylcytidine, 5,2-0-dimethylcytidine, 2-0-methylcytidine, N4,2-
0-dimethylcytidine,
N4,N4,2-0-trimethylcytidine, 1-methylguanosine, N2,7-dimethylguanosine, N2-
methylguanosine, 2-0-
ribosylphosphate guanosine, 7-methylguanosine, under modified
hydroxywybutosine, 7-aminomethy1-
7-deazaguanosine, 7-cyano-7-deazaguanosine, N2,N2-dimethylguanosine, 4-
demethylwyosine,
epoxyqueuosine, hydroxywybutosine, isowyosine, N2,7,2-0-trimethylguanosine,
N2,2-0-
dimethylguanosine, 1,2-0-dimethylguanosine, 2-0-methylguanosine, N2,N2,2-0-
trimethylguanosine,
N2,N2,7-trimethylguanosine, peroxywybutosine, galactosyl-queuosine, mannosyl-
queuosine,
queuosine, archaeosine, wybutosine, methylwyosine, wyosine, 2-thiouridine, 3-
(3-amino-3-
carboxypropyl)uridine, 3-methyluridine, 4-thiouridine, 5-methyl-2-thiouridine,
5-
methylaminomethyluridine, 5-carboxymethyluridine, 5-
carboxymethylaminomethyluridine, 5-
hydroxyuridine, 5-methyluridine, 5-taurinomethyluridine, 5-
carbamoylmethyluridine, 5-
(carboxyhydroxymethyl)uridine methyl ester, dihydrouridine, 5-
methyldihydrouridine, 5-
methylaminomethy1-2-thiouridine, 5-(carboxyhydroxymethyOuridine, 5-
(isopentenylaminomethyl)uridine, 5-(isopentenylaminomethyl)-2-thiouridine, 3,2-
0-dimethyluridine, 5-
carboxymethylaminomethy1-2-0-methyluridine, 5-carbamoylmethy1-2-0-
methyluridine, 5-
methoxycarbonylmethy1-2-0-methyluridine, 5-(isopentenylaminomethyl)-2-0-
methyluridine, 5,2-0-
dimethyluridine, 2-0-methyluridine, 2-thio-2-0-methyluridine, uridine 5-
oxyacetic acid, 5-
methoxycarbonylmethyluridine, uridine 5-oxyacetic acid methyl ester, 5-
methoxyuridine, 5-
aminomethy1-2-thiouridine, 5-carboxymethylaminomethy1-2-thiouridine, 5-
methylaminomethy1-2-
selenouridine, 5-methoxycarbonylmethy1-2-thiouridine, 5-taurinomethy1-2-
thiouridine, pseudouridine,
1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine, 1-methylpseudouridine, 3-
methylpseudouridine,
2-0-methylpseudouridine, inosine, 1-methylinosine, 1,2-0-dimethylinosine and 2-
0-methylinosine.
Each of these or the modified nucleobase thereof may be components of nucleic
acids of the present
invention.
The polynucleotides of the invention includes a first region of linked
nucleosides encoding
a polypeptide of interest, a first flanking region located at the 5' terminus
of the first region, and a
second flanking region located at the 3' terminus of the first region.
In some embodiments, the polynucleotide (e.g., the first region, first
flanking region, or
second flanking region) includes n number of linked nucleosides having Formula
(la) or Formula (la-
1):
_______________________________ yl y5
______ y1 y5 B Rt\
R5
/ R2Dr\ mu
R5-7 ( y2 \ R27 ,
M
y2 R27 m, M Y3=13 _______________
y3=ID _________________
I
I I 4
- (la) - (la-1) or a
pharmaceutically
acceptable salt or stereoisomer thereof, wherein U is 0, S, N(RU)nu, or
C(Ru)õ, wherein nu is an
integer from 0 to 2 and each Ru is, independently, H, halo, or optionally
substituted alkyl;
- 22 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
- - - is a single bond or absent;
each of R1, R2, R1, R2, R3, R4, and R5, if present, is, independently, H,
halo, hydroxy,
thiol, optionally substituted alkyl, optionally substituted alkynyl,
optionally substituted alkoxy,
optionally substituted alkenyloxy, optionally substituted alkynyloxy,
optionally substituted
aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted
hydroxyalkoxy, optionally
substituted amino, azido, optionally substituted aryl, optionally substituted
aminoalkyl, optionally
substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent;
wherein R3 with one or more
of R1, R1, R2, R2, or R5 (e.g., R1 and R3, R1 and R3, R2 and R3, R2 and R3, or
R5 and R3) can join
together to form optionally substituted alkylene or optionally substituted
heteroalkylene and, taken
together with the carbons to which they are attached, provide an optionally
substituted heterocyclyl
(e.g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); wherein R5 with
one or more of R1, R1, R2, or R2
(e.g., R1 and R5, R1 and R5, R2' and R5, or R2 and R5) can join together to
form optionally substituted
alkylene or optionally substituted heteroalkylene and, taken together with the
carbons to which they
are attached, provide an optionally substituted heterocyclyl (e.g., a
bicyclic, tricyclic, or tetracyclic
heterocyclyl); and wherein R4 and one or more of R15 R15 R25 R25 R35 or R5 can
join together to form
optionally substituted alkylene or optionally substituted heteroalkylene and,
taken together with the
carbons to which they are attached, provide an optionally substituted
heterocyclyl (e.g., a bicyclic,
tricyclic, or tetracyclic heterocyclyl);
each of m' and m" is, independently, an integer from 0 to 3 (e.g., from 0 to
2, from 0 to 15
from 1 to 3, or from 1 to 2);
each of Y15 Y25 and Y35 is, independently, 0, S5 Se, -NR-, optionally
substituted alkylene,
or optionally substituted heteroalkylene, wherein RN1 is H, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
aryl, or absent;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted
alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
alkoxy, optionally substituted
alkenyloxy, optionally substituted alkynyloxy, optionally substituted
thioalkoxy, optionally substituted
alkoxyalkoxy, or optionally substituted amino;
each Y5 is 0, 55 Se, optionally substituted alkylene (e.g., methylene), or
optionally
substituted heteroalkylene;
n is an integer from 1 to 100,000; and
B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof),
wherein B and R15
B and R25 B and R15 or B and R2 can, taken together with the carbons to which
they are attached,
optionally form a bicyclic group (e.g., a bicyclic heterocyclyl) or wherein B,
R1, and R3 or B, R2, and
R3 can optionally form a tricyclic or tetracyclic group (e.g., a tricyclic or
tetracyclic heterocyclyl, such
as in Formula (11o)-(11p) herein).
In some embodiments, the polynucleotide includes a modified ribose. In some
embodiments, the polynucleotide (e.g., the first region, the first flanking
region, or the second flanking
region) includes n number of linked nucleosides having Formula (1a-2)-(1a-5)
or a pharmaceutically
acceptable salt or stereoisomer thereof.
- 23 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
_______ y1 y5 B __________ y1 y5
'
Y2 my2 \ k2/ M
1 1 \ irrf
Y3=P ___________ Y3=Pi __________
jit4 yl4
¨(la-2), ¨ ¨ (la-3),
_____ y1 y5 ___________________ y1 y5
_ R4 R5
,R4
RI' (
7.
y2
y2 \ M
1 1 \ irrf
N(3=Ri _________ Y3=Pi __________
\!/4 yl4
¨ (la-4), ¨ ¨ (la-5).
In some embodiments, the polynucleotide (e.g., the first region, the first
flanking region, or
the second flanking region) includes n number of linked nucleosides having
Formula (lb) or Formula
(lb-1):
R3" u B
(<1,1
R4
Ra, 1-- - R4
R5 Y2 R5 Y2
Y3 Y3=1Di __
yI4 \!/4
¨ (lb), _ _ (lb-1)
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
U is 0, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each
Ru is,
independently, H, halo, or optionally substituted alkyl;
- - - is a single bond or absent;
each of R1, R3, R3, and R4 is, independently, H, halo, hydroxy, optionally
substituted alkyl,
optionally substituted alkoxy, optionally substituted alkenyloxy, optionally
substituted alkynyloxy,
optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy,
optionally substituted
hydroxyalkoxy, optionally substituted amino, azido, optionally substituted
aryl, optionally substituted
aminoalkyl, optionally substituted aminoalkenyl, optionally substituted
aminoalkynyl, or absent; and
wherein R1 and R3 or R1 and R3 can be taken together to form optionally
substituted alkylene or
optionally substituted heteroalkylene (e.g., to produce a locked nucleic
acid);
each R5 is, independently, H, halo, hydroxy, optionally substituted alkyl,
optionally
substituted alkoxy, optionally substituted alkenyloxy, optionally substituted
alkynyloxy, optionally
substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent;
- 24 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of Y1, Y2, and Y3 is, independently, 0, S, Se, NR-, optionally
substituted alkylene,
or optionally substituted heteroalkylene, wherein RN1 is H, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, or optionally substituted
aryl;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted
alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
alkoxy, optionally substituted
alkenyloxy, optionally substituted alkynyloxy, optionally substituted
alkoxyalkoxy, or optionally
substituted amino;
n is an integer from 1 to 100,000; and
B is a nucleobase.
In some embodiments, the polynucleotide (e.g., the first region, first
flanking region, or
second flanking region) includes n number of linked nucleosides having Formula
(lc):
______ y1 _y5 u B3
Rb3
R5 B' B2
-
y2 01
y3=Pi __________________
NI(4
¨
(lc), or a pharmaceutically acceptable salt or stereoisomer thereof,
wherein
U is 0, S, N(RU)nu, or C(Ru)õ, wherein nu is an integer from 0 to 2 and each
Ru is,
independently, H, halo, or optionally substituted alkyl;
- - - is a single bond or absent;
each of B1, B2, and B3 is, independently, a nucleobase (e.g., a purine, a
pyrimidine, or
derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally
substituted alkyl, optionally
substituted alkoxy, optionally substituted alkenyloxy, optionally substituted
alkynyloxy, optionally
substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally
substituted hydroxyalkoxy,
optionally substituted amino, azido, optionally substituted aryl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl,
wherein one and only one
of B1, B2, and B3 is a nucleobase;
each of Rbl, Rb2, Rb3, R3, and R5 is, independently, H, halo, hydroxy, thiol,
optionally
substituted alkyl, optionally substituted alkoxy, optionally substituted
alkenyloxy, optionally substituted
alkynyloxy, optionally substituted aminoalkoxy, optionally substituted
alkoxyalkoxy, optionally
substituted hydroxyalkoxy, optionally substituted amino, azido, optionally
substituted aryl, optionally
substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally
substituted aminoalkynyl;
each of Y1, Y2, and Y3, is, independently, 0, S, Se, -NR-, optionally
substituted alkylene,
or optionally substituted heteroalkylene, wherein RN1 is H, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, or optionally substituted
aryl;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted
alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
alkoxy, optionally substituted
- 25 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
alkenyloxy, optionally substituted alkynyloxy, optionally substituted
thioalkoxy, optionally substituted
alkoxyalkoxy, or optionally substituted amino;
each Y5 is 0, S, Se, optionally substituted alkylene (e.g., methylene), or
optionally
substituted heteroalkylene;
n is an integer from 1 to 100,000; and
wherein the ring including U can include one or more double bonds.
In particular embodiments, the ring including U does not have a double bond
between U-
CB3Rb3 or between CB3Rb3-cB2Rb2.
In some embodiments, the polynucleotide (e.g., the first region, first
flanking region, or
second flanking region) includes n number of linked nucleosides having Formula
(Id):
_______ 1 5 B
Y Y
R3
y2
Y34 ______________
¨ (Id), or a pharmaceutically acceptable salt or
stereoisomer thereof, wherein U is
0, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each Ru
is, independently, H, halo,
or optionally substituted alkyl;
each R3 is, independently, H, halo, hydroxy, thiol, optionally substituted
alkyl, optionally
substituted alkoxy, optionally substituted alkenyloxy, optionally substituted
alkynyloxy, optionally
substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally
substituted hydroxyalkoxy,
optionally substituted amino, azido, optionally substituted aryl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
each of Y1, Y2, and Y3, is, independently, 0, S, Se, -NR-, optionally
substituted alkylene,
or optionally substituted heteroalkylene, wherein RN1 is H, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, or optionally substituted
aryl;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted
alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
alkoxy, optionally substituted
alkenyloxy, optionally substituted alkynyloxy, optionally substituted
thioalkoxy, optionally substituted
alkoxyalkoxy, or optionally substituted amino;
each Y5 is 0, S, optionally substituted alkylene (e.g., methylene), or
optionally substituted
heteroalkylene;
n is an integer from 1 to 100,000; and
B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
In some embodiments, the polynucleotide (e.g., the first region, first
flanking region, or
second flanking region) includes n number of linked nucleosides having Formula
(le):
- 26 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
U'
_______ Y5
\NB
R6 ¨N ______________
- (le), or a pharmaceutically acceptable salt or
stereoisomer thereof,
wherein each of U' and U" is, independently, 0, S, N(RU)nu, or C(RU)nu,
wherein nu is an
integer from 0 to 2 and each Ru is, independently, H, halo, or optionally
substituted alkyl;
each R6 is, independently, H, halo, hydroxy, thiol, optionally substituted
alkyl, optionally
substituted alkoxy, optionally substituted alkenyloxy, optionally substituted
alkynyloxy, optionally
substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally
substituted hydroxyalkoxy,
optionally substituted amino, azido, optionally substituted aryl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
each Y5' is, 0, S5 optionally substituted alkylene (e.g., methylene or
ethylene), or
optionally substituted heteroalkylene;
n is an integer from 1 to 100,000; and
B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
In some embodiments, the polynucleotide (e.g., the first region, first
flanking region, or
second flanking region) includes n number of linked nucleosides having Formula
(If) or (If-1):
______ y1_y5 B __________ y1_y5 B
R3-
R2" \R2.. R2/ \R2"
y2 y2
1 1
y3=Pi _____________________________ y3=P1 _______
yl4 yl4
¨ ¨ ¨ (If-1), or a pharmaceutically
acceptable salt or stereoisomer thereof,
wherein each of U' and U" is, independently, 0, S, N, N(Ru)õ, or C(Ru)õ,
wherein nu is an
integer from 0 to 2 and each Ru is, independently, H, halo, or optionally
substituted alkyl (e.g., U' is 0
and U" is N);
- - - is a single bond or absent;
each of R1, R2, R1, R2, R35 and R4 is, independently, H, halo, hydroxy, thiol,
optionally
substituted alkyl, optionally substituted alkoxy, optionally substituted
alkenyloxy, optionally substituted
alkynyloxy, optionally substituted aminoalkoxy, optionally substituted
alkoxyalkoxy, optionally
substituted hydroxyalkoxy, optionally substituted amino, azido, optionally
substituted aryl, optionally
substituted aminoalkyl, optionally substituted aminoalkenyl, optionally
substituted aminoalkynyl, or
absent; and wherein R1' and R3, R1" and R3, R2' and R3, or R2" and R3 can be
taken together to form
optionally substituted alkylene or optionally substituted heteroalkylene
(e.g., to produce a locked
nucleic acid); each of m' and m" is, independently, an integer from 0 to 3
(e.g., from 0 to 2, from 0 to
1, from 1 to 3, or from 1 to 2);
- 27 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of Y1, Y2, and Y3, is, independently, 0, S, Se, -NR-, optionally
substituted alkylene,
or optionally substituted heteroalkylene, wherein RN1 is H, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
aryl, or absent;
each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted
alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
alkoxy, optionally substituted
alkenyloxy, optionally substituted alkynyloxy, optionally substituted
thioalkoxy, optionally substituted
alkoxyalkoxy, or optionally substituted amino;
each Y5 is 0, S, Se, optionally substituted alkylene (e.g., methylene), or
optionally
substituted heteroalkylene;
n is an integer from 1 to 100,000; and
B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof).
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), the ring including U has
one or two double bonds.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), each of R1, R1, and R1, if
present, is H. In further embodiments, each of R2, R2, and R2, if present, is,
independently, H, halo
(e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or
ethoxy), or optionally substituted
alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is -(C1-12)s2(OCH2C1-
12)s1 (C1-12)s3OR', wherein
51 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2
and s3, independently, is an
integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to
6, or from 1 to 10), and R' is
H or C1_20 alkyl). In some embodiments, s2 is 0, s1 is 1 or 2, s3 is 0 or 1,
and R' is C1_6 alkyl.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(10, (11a)-(11p),
(11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and (IXa)-
(1Xr)), each of R2, R2, and R2, if
present, is H. In further embodiments, each of R1, R1, and R1, if present, is,
independently, H, halo
(e.g., fluoro), hydroxy, optionally substituted alkoxy (e.g., methoxy or
ethoxy), or optionally substituted
alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is -(C1-12)s2(OCH2C1-
12)s1 (C1-12)s3OR', wherein
51 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2
and s3, independently, is an
integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to
6, or from 1 to 10), and R' is
H or C1_20 alkyl). In some embodiments, s2 is 0, s1 is 1 or 2, s3 is 0 or 1,
and R' is C1_6 alkyl.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), each of R3, R4, and R5 is,
independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl,
optionally substituted alkoxy
(e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In
particular embodiments, R3 is H,
R4 is H, R5 is H, or R3, R4, and R5 are all H. In particular embodiments, R3
is C1_6 alkyl, R4 is C1_6
alkyl, R5 is C1_6 alkyl, or R3, R4, and R5 are all C1_6 alkyl. In particular
embodiments, R3 and R4 are
both H, and R5 is C1_6 alkyl.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), R3 and R5 join together to
form optionally substituted alkylene or optionally substituted heteroalkylene
and, taken together with
the carbons to which they are attached, provide an optionally substituted
heterocyclyl (e.g., a bicyclic,
- 28 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
tricyclic, or tetracyclic heterocyclyl, such as trans-3',4' analogs, wherein
R3 and R5 join together to
form heteroalkylene (e.g., -(C1-12)biO(C1-12)b20(C1-12)b3-, wherein each of
b1, b2, and b3 are,
independently, an integer from 0 to 3).
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), R3 and one or more of
R1, R1, R2', R2, or R5join together to form optionally substituted alkylene or
optionally substituted
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic
heterocyclyl, R3 and one or more of
R1, R1, R2', R2, or R5 join together to form heteroalkylene (e.g., -(C1-
12)biO(C1-12)b20(C1-12)b3-, wherein
each of b1, b2, and b3 are, independently, an integer from 0 to 3).
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), R5 and one or more of
R1, R1, R2', or R2 join together to form optionally substituted alkylene or
optionally substituted
heteroalkylene and, taken together with the carbons to which they are
attached, provide an optionally
substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic
heterocyclyl, R5 and one or more of
R1, R1, R2', or R2 join together to form heteroalkylene (e.g., -(C1-12)biO(C1-
12)b20(C1-12)b3-, wherein each
of b1, b2, and b3 are, independently, an integer from 0 to 3).
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), each Y2 is,
independently, 0, S, or -NR-, wherein RN1 is H, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, or optionally substituted aryl. In
particular embodiments, Y2 is
NR-, wherein RN1 is H or optionally substituted alkyl (e.g., C1_6 alkyl, such
as methyl, ethyl,
isopropyl, or n-propyl).
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), each Y3 is,
independently, 0 or S.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), R1 is H; each R2 is,
independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy
(e.g., methoxy or ethoxy),
or optionally substituted alkoxyalkoxy (e.g., -(CH2)s2(OCH2CH2)s1(CH2)s3OR',
wherein 51 is an integer
from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer from 0 to
10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to
10), and R' is H or C1_20 alkyl,
such as wherein s2 is 0, 51 is 1 or 2, s3 is 0 or 1, and R' is C1_6 alkyl);
each Y2 is, independently, 0 or
-NR-, wherein RN1 is H, optionally substituted alkyl, optionally substituted
alkenyl, optionally
substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or
optionally substituted alkyl
(e.g., C1_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each
Y3 is, independently, 0 or S
(e.g., S). In further embodiments, R3 is H, halo (e.g., fluoro), hydroxy,
optionally substituted alkyl,
optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally
substituted alkoxyalkoxy. In yet
further embodiments, each Y1 is, independently, 0 or -NR-, wherein RN1 is H,
optionally substituted
alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or
optionally substituted aryl (e.g.,
wherein RN1 is H or optionally substituted alkyl (e.g., C1_6 alkyl, such as
methyl, ethyl, isopropyl, or n-
- 29 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally
substituted alkyl, optionally
substituted alkoxy, optionally substituted thioalkoxy, optionally substituted
alkoxyalkoxy, or optionally
substituted amino.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), each R1 is,
independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkoxy
(e.g., methoxy or ethoxy),
or optionally substituted alkoxyalkoxy (e.g., -(CH2),2(OCH2CH2),1(CH2),30R',
wherein 51 is an integer
from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer from 0 to
(e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10),
and R' is H or C1_20 alkyl,
10 such as wherein s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R' is C1_6
alkyl); R2 is H; each Y2 is,
independently, 0 or -NR-, wherein RN1 is H, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g.,
wherein RN1 is H or
optionally substituted alkyl (e.g., C1_6 alkyl, such as methyl, ethyl,
isopropyl, or n-propyl)); and each Y3
is, independently, 0 or S (e.g., S). In further embodiments, R3 is H, halo
(e.g., fluoro), hydroxy,
optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or
ethoxy), or optionally
substituted alkoxyalkoxy. In yet further embodiments, each Y1 is ,
independently, 0 or -NR-,
wherein RN1 is H, optionally substituted alkyl, optionally substituted
alkenyl, optionally substituted
alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally
substituted alkyl (e.g., C1_6
alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is,
independently, H, hydroxy, thiol,
optionally substituted alkyl, optionally substituted alkoxy, optionally
substituted thioalkoxy, optionally
substituted alkoxyalkoxy, or optionally substituted amino.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), the ring including U is in
the 13-D (e.g., 13-D-ribo) configuration.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), the ring including U is in
the a-L (e.g., a-L-ribo) configuration.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), one or more B is not
pseudouridine (t.p) or 5-methyl-cytidine (m5C).
In some embodiments, about 10% to about 100% of n number of B nucleobases is
not 4)
or m5C (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to
60%, from 10% to
75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from
20% to 35%,
from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20%
to 95%, from
20% to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to
75%, from 50% to
90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from
75% to 90%,
from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% of n
number of B is
not 4) or m5C). In some embodiments, B is not 4) or m5C.
In some embodiments of the polynucleotides (e.g., Formulas (1a)-(1a-5), (1b)-
(1f-1), (11a)-
(11p), (11b-1), (11b-2), (11c-1)-(11c-2), (11n-1), (11n-2), (1Va)-(IVI), and
(IXa)-(1Xr)), when B is an unmodified
- 30 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
nucleobase selected from cytosine, guanine, uracil and adenine, then at least
one of Y1, Y2, or Y3 is
not 0.
In some embodiments, the polynucleotide includes a modified ribose. In some
embodiments, the polynucleotide (e.g., the first region, the first flanking
region, or the second flanking
region) includes n number of linked nucleosides having Formula (11a)-(11c):
______________________________ yl_y5 g
________ Y1 Y5
11
___________________________________________ R4
R74
R R2 R y2 R2
Y2 -
Y3=R ___________________________________________
Y3=P __________________
\!14
4
¨ . (11a), ¨ (11b), or
________ yl y5
,U
\R5 11
R3 _______________ R4
R2
y2
Y3 =P _________________
yl4
- lc), or a pharmaceutically acceptable salt or
stereoisomer thereof. In
particular embodiments, U is 0 or C(RU)nu, wherein nu is an integer from 0 to
2 and each Ru is,
independently, H, halo, or optionally substituted alkyl (e.g., U is ¨CH2¨ or
¨CH¨). In other
embodiments, each of R1, R2, R3, R4, and R5 is, independently, H, halo,
hydroxy, thiol, optionally
substituted alkyl, optionally substituted alkoxy, optionally substituted
alkenyloxy, optionally substituted
alkynyloxy, optionally substituted aminoalkoxy, optionally substituted
alkoxyalkoxy, optionally
substituted hydroxyalkoxy, optionally substituted amino, azido, optionally
substituted aryl, optionally
substituted aminoalkyl, optionally substituted aminoalkenyl, optionally
substituted aminoalkynyl, or
absent (e.g., each R1 and R2 is, independently H, halo, hydroxy, optionally
substituted alkyl, or
optionally substituted alkoxy; each R3 and R4 is, independently, H or
optionally substituted alkyl; and
R5 is H or hydroxy), and - - - is a single bond or double bond.
In particular embodiments, the polynucleotide (e.g., the first region, the
first flanking
region, or the second flanking region) includes n number of linked nucleosides
having Formula (11b-1)-
(11b-2):
- 31 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
yi_ y5 u g y1¨y5
y2R2 y2 R2
1 1
Y3lp _______________ y3=Ri ____
\!/4 yl4
(11b-1) or ¨ (11b-2) or a pharmaceutically
acceptable
salt or stereoisomer thereof. In some embodiments, U is 0 or C(Ru)õ, wherein
nu is an integer from
0 to 2 and each Ru is, independently, H, halo, or optionally substituted alkyl
(e.g., U is ¨CH2¨ or ¨
CH). In other embodiments, each of R1, R2, and R2 is, independently, H, halo,
hydroxy, thiol,
optionally substituted alkyl, optionally substituted alkoxy, optionally
substituted alkenyloxy, optionally
substituted alkynyloxy, optionally substituted aminoalkoxy, optionally
substituted alkoxyalkoxy,
optionally substituted hydroxyalkoxy, optionally substituted amino, azido,
optionally substituted aryl,
optionally substituted aminoalkyl, optionally substituted aminoalkenyl,
optionally substituted
aminoalkynyl, or absent (e.g., each R1, R2, and R2 is, independently, H, halo,
hydroxy, optionally
substituted alkyl, or optionally substituted alkoxy, e.g., H, halo, hydroxy,
alkyl, or alkoxy). In particular
embodiments, R2 or R2 is hydroxy or optionally substituted alkoxy (e.g.,
methoxy, ethoxy, or any
described herein).
In particular embodiments, the polynucleotide (e.g., the first region, the
first flanking
region, or the second flanking region) includes n number of linked nucleosides
having Formula (11c-1)-
(11c-4):
Y1
_____________ U Y1 Y5 Y5
Ko'11
Y2 rµ Y2 rµ
Y3_ 1
Y3=R ______________ ¨P _______
I 4 1 4
¨ (11c-1), ¨ ¨ (11c-2),
Y1 Y5 U y1 y5 0
R3 ___________________ R3
Y2 rµ Y2 rµ
Y3_ 1
Y3=R _________________ ¨P _______
I 4 1 4
¨ (11c-3), or ¨ (11c-4), or a
pharmaceutically
acceptable salt or stereoisomer thereof.
In some embodiments, U is 0 or C(Ru)õ, wherein nu is an integer from 0 to 2
and each Ru
is, independently, H, halo, or optionally substituted alkyl (e.g., U is ¨CH2¨
or ¨CH¨). In some
embodiments, each of R1, R2, and R3 is, independently, H, halo, hydroxy, thio,
optionally substituted
- 32 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy,
optionally substituted alkynyloxy,
optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy,
optionally substituted
hydroxyalkoxy, optionally substituted amino, azido, optionally substituted
aryl, optionally substituted
aminoalkyl, optionally substituted aminoalkenyl, optionally substituted
aminoalkynyl, or absent (e.g.,
each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted
alkyl, or optionally
substituted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R3 is,
independently, H or
optionally substituted alkyl)). In particular embodiments, R2 is optionally
substituted alkoxy (e.g.,
methoxy or ethoxy, or any described herein). In particular embodiments, R1 is
optionally substituted
alkyl, and R2 is hydroxy. In other embodiments, R1 is hydroxy, and R2 is
optionally substituted alkyl.
In further embodiments, R3 is optionally substituted alkyl.
In some embodiments, the polynucleotide includes an acyclic modified ribose.
In some
embodiments, the polynucleotide (e.g., the first region, the first flanking
region, or the second flanking
region) includes n number of linked nucleosides having Formula (11d)-(110:
____________ Y1 Y5
_______________________________ Y1 Y5
R3 R4 / 4
R3 R
m,2 m,2
Y2 rµ Y2 rµ
Y3=R ____________________________ y3=R _______
4 4
¨ (11d), ¨ (Ile), or
____________ Y1 Y5
\R5
R3 R4
Y2 IA
Y3=R ___________________
4
¨ (110, or a pharmaceutically acceptable salt or stereoisomer thereof,
wherein the variables are as defined above.
In some embodiments, the polynucleotide includes an acyclic modified hexitol.
In some
embodiments, the polynucleotide (e.g., the first region, the first flanking
region, or the second flanking
region) includes n number of linked nucleosides having Formula (11g)-(11j):
____________ y1 y5 B _________ y1 y5
'"R4 R3,:rij"R4
R5
R1" R5: R1/R1
iR2" 'R2"
y2 R2 y2 R2'
Y3=Pi ____________________________ Y3=Pi ___________
I 4 I 4
_ (11g), _ _ (11h),
- 33 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
_______ y1 y5 B3 ____________ y1 y5 B3
R3- R51 B2 R3:11j,,1
' "Rb3
.13' B2 R5 B' B2
y2 01 y2 141D1
1 1
y3=1? ____________________________ y3=1? ___________
I 4 I 4
_ (Ili), or _ _ (11j), or a
pharmaceutically
acceptable salt or stereoisomer thereof.
In some embodiments, the polynucleotide includes a sugar moiety having a
contracted or
an expanded ribose ring. In some embodiments, the polynucleotide (e.g., the
first region, the first
flanking region, or the second flanking region) includes n number of linked
nucleosides having
Formula (11k)-(11m):
_______ y1 y5 B __________ yl y5
U
U
R5 '1
.4
R3 .2 R3 R4
y2 ill y2
y3=R _______________________________ y3=Pi _____
yI4 yI4
¨ (Ilk), _ _ (111), or
_______ y1 y5
R3 v-U,4
'
R5 R R.1"
= R2"
y2 R2'
y31=1? ___________________
1 4
_ (11m), or a pharmaceutically acceptable salt or
stereoisomer thereof,
wherein each of R1, R2, R1, R1, R2, and R2 is, independently, H, halo,
hydroxy, optionally substituted
alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy,
optionally substituted alkynyloxy,
optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or
absent; and wherein R2
and R3 or R2 and R3 can be taken together to form optionally substituted
alkylene or optionally
substituted heteroalkylene.
In some embodiments, the polynucleotide includes a locked modified ribose. In
some
embodiments, the polynucleotide (e.g., the first region, the first flanking
region, or the second flanking
region) includes n number of linked nucleosides having Formula (11n):
- 34 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
____________ Y1 Y5
R1/
/3 R4
Y3=P __________________
I 4
¨ (11n), or a pharmaceutically acceptable salt or stereoisomer thereof,
wherein R3 is 0, S, or -NR-, wherein RN1 is H, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3
is optionally substituted
alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-) or optionally substituted
heteroalkylene (e.g., -
CH2NH-, -CH2CH2NH-, -CH2OCH2-, or -CH2CH2OCH2-) (e.g., R3' is 0 and R3 is
optionally substituted
alkylene (e.g., -CH2-, -CH2CH2-, or -CH2CH2CH2-)).
In some embodiments, the polynucleotide (e.g., the first region, the first
flanking region, or
the second flanking region) includes n number of linked nucleosides having
Formula (11n-1)-(11-n2):
____________ Y1 Y5
________________________________________ Y1_Y5
R13-)
Y2 0
Y3=P _______________________________ y3=P _______
4 4
¨ (11n-1) or ¨ (11n-2), or a pharmaceutically
acceptable salt or stereoisomer thereof, wherein R3 is 0, S, or -NR-, wherein
RN1 is H, optionally
substituted alkyl, optionally substituted alkenyl, optionally substituted
alkynyl, or optionally substituted
aryl and R3 is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -
CH2CH2CH2-) or optionally
substituted heteroalkylene (e.g., -CH2NH-, -CH2CH2NH-, -CH2OCH2-, or -
CH2CH2OCH2-) (e.g., R3 is
0 and R3 is optionally substituted alkylene (e.g., -CH2-, -CH2CH2-, or -
CH2CH2CH2-)).
In some embodiments, the polynucleotide includes a locked modified ribose that
forms a
tetracyclic heterocyclyl. In some embodiments, the polynucleotide (e.g., the
first region, the first
flanking region, or the second flanking region) includes n number of linked
nucleosides having
Formula (110):
____________ Y1 Y5
R4 _______________ v1 v5 R4
\ T2' ,R12a
T2' ,R12a
R3 (N/r2" Ncrl __ / N
¨ N12c
R __________________
3
2 V - __ -V3
T.1"
3_ _____________________________________________________________
Y3=P __________________________________________ Y ¨P
\fr 4
¨ (11o) or ¨ ¨ (11p), or
a
pharmaceutically acceptable salt or stereoisomer thereof, wherein the dashed
lines represent optional
double bonds, and wherein R12a, R12c5 T15 Ti 5T25 T25 ¨15
v and V3 are as described herein.
- 35 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Any of the formulas for the polynucleotides can include one or more
nucleobases
described herein.
In one embodiment, the present invention provides methods of preparing a
polynucleotide
comprising at least one nucleotide, wherein the polynucleotide comprises n
number of nucleosides
having Formula (la), as defined herein:
_______ y1 y5
N ,, 4
U/
R3" R1"
OR2"
y2 \ R
2
/m'
Y3=Pi _____________________
I 4
_ (la),
the method comprising reacting a compound of Formula (111a), as defined
herein:
/y3\
y6 _____ p y1 __ y5
\'4 /
Ro R '\
"
y3=p _______________ y6
\ xi14/
/q
(111a),
with an RNA polymerase, and a cDNA template.
In one embodiment, the present invention provides methods of preparing a
polynucleotide
comprising at least one nucleotide, wherein the polynucleotide comprises n
number of nucleosides
having Formula (1a-1):
________ yl_y5
/
õ
KR2 mil
y2 R27
y4
¨ (la-1), the method comprising reacting a
compound of
Formula (111a-1), as defined herein:
- 36 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
y3 \
y6 _____ p yl ___ y5
NI
\U (4 A 4
R5,7 Rfr
y2 D-
y3=p ______________ y6
\ v4/
(111a-1), with an RNA polymerase, and a cDNA template.
In a further embodiment, the present invention provides methods of amplifying
a
polynucleotide comprising at least one nucleotide (e.g., modified mRNA
molecule), the method
comprising: reacting a compound of Formula (111a-1), as defined herein, with a
primer, a cDNA
template, and an RNA polymerase.
In one embodiment, the present invention provides methods of preparing a
polynucleotide
comprising at least one nucleotide, wherein the polynucleotide comprises n
number of nucleosides
having Formula (1a-2), as defined herein:
______________________ yl y5 U B
R3 - R4
2'
/
y2 ril
Y3=Pi __
+4
¨
(1a-2), the method comprising reacting a compound of Formula (Ma-
i() 2), as defined herein:
7 c3
,
y6 ______________ p y ys,
\ y4 ir 4
R3 ¨
2t
T2
y313 __ y6
4 /
'q
(111a-2), with an RNA polymerase, and a cDNA template.
In a further embodiment, the present invention provides methods of amplifying
a
polynucleotide comprising at least one nucleotide (e.g., modified mRNA
molecule), the method
comprising reacting a compound of Formula (111a-2), as defined herein, with a
primer, a cDNA
template, and an RNA polymerase.
In some embodiments, the reaction may be repeated from 1 to about 7,000 times.
In any
of the embodiments herein, B may be any nucleobase provided herein.
- 37 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
The polynucleotides can optionally include 5' and/or 3' flanking regions,
which are
described herein.
Modified Nucleotides and Nucleosides
The present invention also includes the building blocks, e.g., modified
ribonucleosides,
modified ribonucleotides, of the polynucleotides, e.g., modified RNA ( or
mRNA) molecules. For
example, these building blocks can be useful for preparing the polynucleotides
of the invention.
In some embodiments, the building block molecule has Formula (111a) or (111a-
1):
/y3 \ /y3 \
I I I I
y6 ______ p y1 __ y5 y6 __ p y1 __ y5
" B
\'4 / \'' sow
R5 RI t
R R R5,1 RI RiN'
s's -
y2 ; 2' y2\\ "DP2'
y3= p _______________ y6 y3= p __ y6
\ /
y4 /
/
(111a), \ (111a-1) or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein the
substituents are as described
herein (e.g., for Formula (la) and (la-1)), and wherein when B is an
unmodified nucleobase selected
from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3
is not 0.
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide, has Formula (IVa)-(1Vb):
7y3
y6 _____ i Tg 5 /y3 \
I Yx7N/
r
y6 __________________________________________ p yl 5
\y4
C)C) r
n (IVa) or HO OH (IVb), or a pharmaceutically
acceptable salt or stereoisomer thereof, wherein B is as described herein
(e.g., any one of (b1)-
(b43)).
In particular embodiments, Formula (IVa) or (IVb) is combined with a modified
uracil (e.g.,
any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula
(b1), (b8), (b28), (b29),
or (b30)). In particular embodiments, Formula (IVa) or (IVb) is combined with
a modified cytosine
(e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as
formula (b10) or (b32)).
In particular embodiments, Formula (IVa) or (IVb) is combined with a modified
guanine (e.g., any one
of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, Formula
(IVa) or (IVb) is
combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and
(b41)-(b43)).
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide, has Formula (IVc)-(1Vk):
- 38 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
( ______ y3 \ ( y3 \
y6 I I
y6 ii:ij_yliy5 B
1=1)¨Y1 r------ y5 p
y4 r v,UNI y4 /r
W _
Ho I-R2 (IVc), HO 12(1Vd),
/ y3 \ 7 y3
1 1 y6 __ ig yl 5
Y6 ______ PI Y1,------. 5 I
7U
\ r V
/1 y4
R3Y\ R3µ
HO R2(IVe), HO R2avo,
7 y3 7 y3 \
I I
y6 ______ p yl,______ 5 I I
I y6 __ p yl
\4 irR3k._ U/3_ RI\
R3 \ A _____________________________________________________ R
HO R2 jrn
¨(IVg), HO OCH3(1Vh),
( 7 y3 \
Y3\
y6 ______ ig y1 5 y6 __ ig y1
I \S5,U 11
y4 ir Y:U/3 \ NI(4 /r
R3 \ RI R3 \
_
HO F (Ivi), HO OCH3(lvi),
(
Y3 \ (3\
y6 ______ p¨ y1 5 y6 __
I I U
y4 ir Y:U/3 y4 ,
R3 \ RI RI
Ho ol (IVk), or HO i (IVO,
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as
described herein (e.g.,
any one of (b1)-(b43)).
In particular embodiments, one of Formulas (IVc)-(1Vk) is combined with a
modified uracil
(e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as
formula (b1), (b8), (b28),
(b29), or (b30)).
- 39 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In particular embodiments, one of Formulas (IVc)-(1Vk) is combined with a
modified
cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-
(b36), such as formula (b10)
or (b32)).
In particular embodiments, one of Formulas (IVc)-(1Vk) is combined with a
modified
guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
In particular embodiments, one of Formulas (IVc)-(1Vk) is combined with a
modified
adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
In other embodiments, the building block molecule, which may be incorporated
into a
polynucleotide has Formula (Va) or (Vb):
R29
7 y3 \ ¨N
ii 7 y3 \ V7 \ ---R27
Y6 ______ Pi Yi __ Y6 1 1 B
1 y6 ______ ii:Ly1 I N N
\ y4 4 R3sku R\I I
111 (Va) or y-7 -1-2 (Vb), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein the
variables are as described
herein (e.g., any one of (b1)-(b43)).
In other embodiments, the building block molecule, which may be incorporated
into a
polynucleotide has Formula (IXa)-(1Xd):
Y3 \
Y II:1) yli_____ 5
(
NI(4 /r VO! Y3
Y
( ig y)_________y
1
y4 r 0!
_ - __ --,
HO F axa), HO Br (IXb),
Y3 /y3
:
y6 ( 111) 4 r y y5
NI( Y\y0! y6 __________ ,g_yi y5
\4 r vOl
Ho CI (NO, or Ho -I (IXd), or a
pharmaceutically
acceptable salt or stereoisomer thereof, wherein B is as described herein
(e.g., any one of (b1)-
(b43)).
In particular embodiments, one of Formulas (IXa)-(1Xd) is combined with a
modified uracil
(e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as
formula (b1), (b8), (b28),
(b29), or (b30)). In particular embodiments, one of Formulas (IXa)-(1Xd) is
combined with a modified
cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-
(b36), such as formula (b10)
or (b32)).
- 40 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In particular embodiments, one of Formulas (IXa)-(1Xd) is combined with a
modified
guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
In particular embodiments, one of Formulas (IXa)-(1Xd) is combined with a
modified
adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
In other embodiments, the building block molecule, which may be incorporated
into a
polynucleotide has Formula (IXe)-(1Xg):
y6 ______ p _v1 a y6 __ ig_yl
04,P \ y5
\ BH2 / r \ BH2 r
-
HO R 2 (Ixe), HO 2 (IX , or
6 (
Y P Y1
yl4 r YO7P
HO
(IXg), or a pharmaceutically acceptable salt or stereoisomer thereof,
wherein B is as described herein (e.g., any one of (b1)-(b43)).
In particular embodiments, one of Formulas (IXe)-(1Xg) is combined with a
modified uracil
(e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as
formula (b1), (b8), (b28),
(b29), or (b30)).
In particular embodiments, one of Formulas (IXe)-(1Xg) is combined with a
modified
cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-
(b36), such as formula (b10)
or (b32)).
In particular embodiments, one of Formulas (IXe)-(1Xg) is combined with a
modified
guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)).
In particular embodiments, one of Formulas (IXe)-(1Xg) is combined with a
modified
adenine (e.g., any one of formulas (b18)-(b20) and (b41)-(b43)).
In other embodiments, the building block molecule, which may be incorporated
into a
polynucleotide has Formula (IXh)-(1Xk):
- 41 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
/Y3 \
Y3
y6 ( II:1) 1____ , I I i
Yu _________________________________ P Y' 5
I T Y5 0 P 14 F----VO!
y4 Jr k R-1 \Y /r OH
_
....-,,-_
HO 0 (IXh), HO b H3 (Ixj),
/y3 7 y3 \
y6 ______ ii:Ly1 Y --y
5 Y6 ___________ ig Y1 -y5
\4 r
1 yl I 7-- 0 P
y \ y4
. k
, cH3
H36 _ 7
..
Ho OH (IXj), or HO OH (IXk), or a pharmaceutically
acceptable salt or stereoisomer thereof, wherein B is as described herein
(e.g., any one of (b1)-
(b43)). In particular embodiments, one of Formulas (IXh)-(1Xk) is combined
with a modified uracil
(e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as
formula (b1), (b8), (b28),
(b29), or (b30)). In particular embodiments, one of Formulas (IXh)-(1Xk) is
combined with a modified
cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-
(b36), such as formula (b10)
or (b32)).
In particular embodiments, one of Formulas (IXh)-(1Xk) is combined with a
modified
guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular
embodiments, one of
Formulas (IXh)-(1Xk) is combined with a modified adenine (e.g., any one of
formulas (b18)-(b20) and
(b41)-(b43)).
In other embodiments, the building block molecule, which may be incorporated
into a
polynucleotide has Formula (IXI)-(1Xr):
/0 \ /0 \ /0 \/
HO ______ P 0 ___ P, 0 B HO __ II? 0P, 0),, B
I 1 1 1
/
0)
\OH / \ BH2 AO) \OH / \ CH3
/ r2 \ rl
HO bH(IXI), HO bH(IXm),
/0 ) r2 \ /Se /0 \
HO ______ P 0 ____ P 0 B H04-0 B
I I 1
\OH !r )OH ) \OH /AO)
HO oH(lxn), Ho F axo,
- 42 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
/'9 \
H
HO-PI ¨0
HO-4-0 B
r
_
HO el(lXp), HO broxco, or
H04-0 B
\OH r
HO bCH3(IXr) or a pharmaceutically acceptable salt or
stereoisomer thereof,
wherein each rl and r2 is, independently, an integer from 0 to 5 (e.g., from 0
to 3, from 1 to 3, or from
1 to 5) and B is as described herein (e.g., any one of (b1)-(b43)).
In particular embodiments, one of Formulas (IXI)-(1Xr) is combined with a
modified uracil
(e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as
formula (b1), (b8), (b28),
(b29), or (b30)).
In particular embodiments, one of Formulas (IXI)-(1Xr) is combined with a
modified
cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-
(b36), such as formula (b10)
or (b32)).
In particular embodiments, one of Formulas (IXI)-(1Xr) is combined with a
modified
guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular
embodiments, one of
Formulas (IXI)-(1Xr) is combined with a modified adenine (e.g., any one of
formulas (b18)-(b20) and
(b41)-(b43)).
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide can be selected from the group consisting of:
NH2
NH
I N-
N N N3
0 \ N N
Ho-(\ri¨o NH2 H00
1
I 0/ \OH /r
H
Ho OH (BB- 1), Ho OH (BB- 2),
C
NH I
\
N
HO 1 NN N HOH=1) 0
=1) __________ f)!
\OH ir
Ho OH (BB- 3), H6 6H (BB- 4),
- 43 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
NH 0
0 1 )
N
HO P 0 0 r\r
i
( ___________
OH /r \
H0,-1' 0 1 ,J
\OI H ir V----1\1
Ho OH (BB- 5), Ho OH (BB- 6),
H3C...,_
U
NH2
/ I 1
HO
(9 \ N---N-
-
_(9 __
HO Fi) 0 \oNiNN NH2
-0
OH /
r r
_ _________________ I
Ho OH (BB-7), HO OH (BB-8),
0 0
N NCH2 H
-(
HO-0 N NH2 HO-O
0 \
OH 1
/r, \oiN NNH
fo\ \ 0 ________________________________________________ ( 1
Pi A ,
OH
, II\INNH2
õ
HO OH (BB- 9), HO OH
(BB- 10),
CI 0
NN
HO-FIL(>-\0 NN NH2
H&-II=LO
\ NNNH2
1 Oi
OH /r \OH
HO OH (BB- 11), and
Ho OH (BB- 12),
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r
is, independently, an
integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
15
- 44 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide can be selected from the group consisting of:
H2N
e
N"--
/0 \
N
, o!
N N H04
H04-0
-0 0
1
\OH /r. \
HO OH (BB- 13), Ho OH (BB- 14),
e e
0 S
\e \ic),)
7o\ 1 ,j /0 )__\
N ---K r
HO lliLO 0 ¨ HO ¨O N N
I OiOH /IA / \OH r
Ho OH (BB- 15), Ha OH (BB- 16),
NH2 G
N------"7"----,N0
-
(
HO-0
1?ii \ n 1 N
OH
r ___
HO OH (BB-17),
- 45 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
H,NvzN0yvNNH2 e
si
N /0 NH-......."-N )_õ\zNiN--
-
/0 \ 1 HO-LP-0 0
1
N \OH
1-101-0A07 N r
õ
9H /1. õ
0- 0-
. ______________ /.
Ho OH (BB-18), (BB-
0
eNH
HO P, -0 N 0
-(
1
OH r
:.- -...
19), and Ho 0H (BB- 20), or a pharmaceutically acceptable
salt or
stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5
(e.g., from 0 to 3, from
1 to 3, or from 1 to 5) and s1 is as described herein.
In some embodiments, the building block molecule, which may be incorporated
into a
nucleic acid (e.g., RNA, mRNA, polynucleotide), is a modified uridine (e.g.,
selected from the group
consisting of:
0 0
H3C HO
/ y3 \ 1 NH
I / y3 \ 1 NH
II NO Nci
y6 _____ p _y1 y6 __ 11:i y1
1 1
y -1.---.0
\ s 4 /r. \ y4 ---'-'r 'VD
FK5 oFi (BB- 21), HO OH (BB- 22),
0
I
7y3 1 NH
y6 _____ p _y1 N '0
NI(4 C)
\ r __
HO OH (BB- 23),
- 46 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
0
H2NNH
(y3 /y3 \ NH
N 0
y6 ____ ig y1y6 ___________________________ 11:i y1 N
\(:)
NI(4 0) , NI(4 O
r _______________________________________ r
= -,.,
HO OH (BB- 24), HO OH (BB- 25),
0 S
/y3 \ NH
7 Y3 NH
y6 ____ ig N,1 N 0 y6 ______ 11:i y1 N(:)
I 1
\y4 --1--''
\ yi 4 C)
r ________________________________________ r __
HO OH (BB- 26), HO OH (BB- 27),
0 0
7 y3 \ HN NH /y3 \ rNH
y6 ____ ig _N/1 (:)
y6 ___ II:i_yl N'N(:)
I 1
\ y4 / r-A0 N!,41 AO)
\ r __
HO OH (BB- 28), Ho 0H (BB- 29),
0
0
------.. --CH3
H3C , HN N
N NH /y3 \
/Y3\ y6 ____ v
ig 1 0
y6 ____ ig _y1 I 1
\
4 0 \ y4 ---L1-. 0 NI(
\
r ________________
HO OH (BB- 30), Ho 0H (BB- 31),
0
F3CNH
1 ? 0 ?,
v6 __ (y3
ig _v1 N O HN OCH3
1 1 1 /y3 \
y4 C) y6 0 1\1
i y1 )
r \4 irA0
Ho OH (BB- 32), HO OH (BB- 33),
- 47 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
0
)-(0CH3HN )c7
HN 1 NH2
Y3 r., 1,1
! 0 Y3
y6_(Vyi 0 N y6_(P_yi - -
1
y4 0) y4 0)
r r
z ____________ :
HO OH (BB- 34), HO OH (BB- 35),
0 0 0
HN)- )cv
1 N CF3 HN 1 NH2
/y3 I H /
0-' -N Y3 NI
y6I_Vyl y6 "" -4_yl
\ y4 0,) \y4 0)
r r
HO OH (BB- 36), Ho OH (BB- 37),
0 0 0
HN N CF3 HN)c7 )cv ,CH3
1 1 hl
L
/y3 S N I H /y3 )1\1
'
y64 _yl y6 _p_Niil
\ y4 0) \ y4 0)
r r
HO OH (BB- 38), Ho OH (BB- 39),
0
0
)czNõCH3
HN 1 HN)NThro
y6
_(
\.
y3 \ ONi 0CF3
11)_yl
NI(4 AO)
/r 1
Y3 ,-,N! oic.,F 0
y6 i, yi - _ _. 3
y4 0)
r
Ho OH (BB- 40), HO OH (BB- 41),
0
0
)cy OH
HN 1 N
HN N ThrOH
Y3 I H
0 )-7
_(Y3 yl )N 'I 0
y4 0
y6_(Vyl 0 N
y6 V S )
) .4 0
r r
z ____________ :
HO OH (BB- 42), HO OH (BB- 43),
0 0 ___________________________
11.õ----.N .,---------õ,OH
y3 HN N
HN )- Thr OFmoc
/y3 j H 0 I H
1 1 1 0 N 0
Y6-HD-Y y6_( yi 0 N
1
\Y4
r r
HO OCH3 (BB- 44), HO OH (BB-
45),
- 48 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0 0
HN N OFmoc HN N)cv OFmoc
)t''V''------'-
Y3 j H
0 Y3 j H 0
y6 _(11)_yl S N y6_(vyi 0 N
NI(4 0) y4 0)
r r
Ho 01H (BB-46), Ha OCH3 (BB- 47),
0 CO2H
...õ...--..... ,..----
...............,
N NH2
O co2Frnoc /y3 1
3
)-N NHFmoc y6
Y I I A
y6 yl N 0 \r+ 0)
1
y4 0) r
r
Ho ^ OH (BB- 48), HO OH (BB- 49),
0 0 0 0
HN HN
)c700Fmoc )c700H
y j
y6
(3 N
1\l
0)
)) 1
y4 j
y_R 1,_yl
y4 0
r
HO ^ OH (BB- 50), Ha OH (BB- 51),
O OFmoc
0 OH
OFmoc
HN 1 _( HN OH
Y3 ON i 0 y3
y6 11. _yl
1
y4 0)
r 1
i
y6 ILI _yi 0 N 0
y4 0)r
HO OH (BB- 52), HO OH (BB- 53),
o OCOCF 3
)-0 Me
HN 1
0 OH
7 Y3 / 0oMe
, I I 1 0 N HN 1
yv _____ p_y . /y3
y6 ON 0
\ NI(L1 0) 4 _yl
1
r \y4 0)
r
-__
HO OH (BB- 54), HO OH (BB- 55),
O 0
y61
OMe OMe
HN 1 HN 1
_(113 1C<ON /y3 yl -
rd=N i 0
y6 yl 4 _
y4 0) \ y4 0)
r r
HO OH (BB-56), I-10 OCH3 (BB-57),
- 49 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0 0
OMe
HN)CZN-CH3
HN 1
I /y3 H
/Y3
y6yi S N 0
y6_11_,(1 0- -N
\ y4 AA0 0/4
HO OH (BB- 58), HO OH (BB- 59),
0
HN N)- -CH
3 0
7y3 j j
HN)C7 -CH
1 N 3
S' N H
y6 ___ iii)_ y1 Y3 Sel\J "
\ y4 0) y6 p _yl
y4 o
r r )
HO OH (BB- 60), HO OH (BB- 61),
O 0
HN 1 HN
NH2 NH2
(_) m 1
7 Y3 i 0 /y3 \
- 0 I\1 0
y6 ___________ P _y1 s"' ' ' y6 iii)¨ y1
\ y4 0) \ y4
r
-
HO OH (BB- 62), Ho 0CH3 (BB- 63),
O CO2Fmoc 0 CO2H
H3C. H3C.NNNH2
N N NHFmoc
/y3 \ /y3 \
y6 y1 r0 y6 __ P _y1
HO OH (BB- 64), HO OH (BB- 65),
O 0
)-OH OFmoc
HN 1 HN I
/ y 3 \0 N / y 3 \ 0Ni 0
ii -
y6 ___ p y1 y6 1:_y1
\.:4 irA0) \ y4 /7'ç'
0)
Ho OH (BB- 66), HO OH (BB- 67),
- 50 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
O 0
)c,
HN N
HN)N,-
1
/y3 I-1 /y3 \ j H
y6
0 N y6 p_y1 S N
__________ yl
\4 ;TV))\ NI(4 1;70
Ho OH (BB- 68), Ho b1-1 (BB-
69),
0
HN)czN,-
/y3 \ j H
y6 ___ il 0 N
y4 irTy)
Ho oCH3 (BB- 70),
O 0
HN A N.,------õ,,
H3C CH3
'NN-
/y3 /y3
y6 ________ _yl r0 y6 yl ---..
\y4 0 \Y4
r _____________
HO OH (BB- 71), HO OH (BB- 72),
O 0
HN AN HN --U-,.N ------.õ..-----
/y3 \ /y3 \
Olr 0
y6 ___ (j y1 y6 __ (j y1
y4 /rA0 \Y4 /174
HO OH (BB- 73), HO OH (BB- 74),
0 0
HN
AN HNAN
7y3 \ nlr iy3 \ ..).,.......,j-
y6 PH -y1 0
y6 ___ ilj_yl s'
0/4 /A,0
HO OH (BB- 75), Ho ol-1 (BB- 76),
- 51 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0 0
HNN HNANOH
A(3 \o /y3
()\
y6 ___ ilj y1 o
OA / riA,0 Y4 ON
r
HO OH (BB- 77), HO OH (BB-78),
O 0
HNHN,--11...,.."--...õ.õ----
)-
/y3 j /y3 \ j
0 N y6 pii y1 0 N
y6 ___ ii:j y1
\4 r 0) \\I(.4 /ITO
HO OH (BB- 79), Ho ol-1 (BB- 80),
0 0
HN HN
/y3 \ ! /y3\ j
0 N ii 0 N
y6 ______ y1
\\
y1
y6 ___________________________________ p _yl
\ y4 A-A,0) \Y4 /17"\,0)
HO OH (BB- 81), HO OH (BB- 82),
O 0
HN HN)-OH
/Y3 \ /y3 Ij
0 N j
0 N
y6 ___ ii:j _y1 y6 ___ ilj_yl
VI(4 AA0) \y4 r 0)
HO OH (BB- 83), HO OH (BB- 84),
O 0 0
/Y3
INH /y3 1 7 Y3 INH
I I
Y6 ___ 11='-y1 NO y6 __ ilj_ y1 N 0 __ Y6
1:_y NO
1 1
\\1(4 0 \y4 0
\ Y4 0 r ,-
r i H CH3 r H3t : __ _)_
HO OH (BB- 85), HO CH3 (BB- 86), HO OH
- 52 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
NH o
/ y3 \ 1
1 1 N 0 \ NH
y6 ____________ p y1 / y3 \ 1
\4 I y6 () _y1 1\1 0
-;-----0
\ y4 r)A0)
(BB- 87), HO 0 (BB- 88), HO I (BB- 89),
0 0
7 Y3
)._,,,,
1NH 1 NH
7 y3 \ 1
y6 ___ II:) yl I N 0 y6 __ i_y1 1\1 0
\ y4 0) \Y4
HO CI (BB- 90), HO Br (BB- 91),
0
0
NH
/Y3 NH \ 1 /Y3 1
y6 ___ ij _y1 1\1 0 y6 __ I)-Yi I\J 0
1 4 (:)
\Y4 ik0 \Y
r
r )
HO 1 (BB- 92), HO CH3 (BB- 93),
0 0
/Y3 CH
NH
1 /Y3 1 N - 3
1
y6 ___ II) _ yl N 0 y6 __ P_yl N 0
\\10 (:) \y4 01
r r
HO OCH3 (BB- 94), HO OH (BB- 95),
0 H3C 0 S
H3CONH HNNH
/Y3 1 /y3 \ N-1 /y3 \
y6 ___ P yl NO y6 __ P y1 N 0 y6 __ _y1 0
1
--1-1:0
\y4 \ 0) \ y4
112"' \y4
/
r
HO OH (BB- 96), HO OH (BB- 97), HO OH
- 53 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0 0
/Y3 \ H3C
HNNH 'NNH
Y3 \
y6 ___________ (i4 _y1 y6 0 S y6 ( ii y1 rs
\Y
(BB- 98), HO OH (BB- 99), HO OH (BB- 100),
0
S
A,-....., ,.....¨...,...õ...,S03H
H3C HN N N
IV NHH
/y3 / l i 3 \ 0
y6 ___ ii:ii y'_ ro y6 __ p y i .
\y4 0 vio
r _____________
HO OH (BB- 101), Ho 01-I (BB- 102),
0
HN ANV\NSO3Fmoc
7y3 \ 0 H
y6 ____ )_y1 -
y4 /1A0
HO OH (BB- 103),
0
HN N ,,,v,,,, __,S03H
/y3 \ (:).'N
I H
y6 ___ (j _y1
y4 /1A0)
HO OH (BB-104),
0
)-
HN N
S03Fmoc
1
7y3 (:).'N "
y6 ____ _y1
y4 0)
r
HO OH (BB- 105),
- 54 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0
S 03H
HN N
7y3 \H S H
N
I
y6 __ A _yl
vi(41 AAO)
HO OH (BB-106),
0 0
)- S03Fmoc
HN N INH
I
7y3 SN H y3 \ N 0
j
y6 __ (j yl y6 __ )_y1
y4 0) y4 / rAO)
r
HO OH (BB- 107), HO OH (BB- 108),
0 0 0
).z),
HN 1 OCH3 HN 1 NH2
/y3
0 N 7y3 \ ON
y6 __ ig yl y6 (j _y 1
\ NI(4 irA0) \Y4 /1A0)
Ho -01-1 (BB- 109), HO OH (BB-
110),
- 55 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0 0 0
0
/y3 H3C NH HN 1 OCH3
1 /y3 01\1
II 1
Y6 ____ Fi'Y NS y6 __ Fi)_y
\ y4 0) \\4 /17\.,0)
r
HO OH (BB- 111), HO OH (BB- 112),
O ? 0
HN
)--0.,.,,CH3
N,..CH3
1
/y3 0
y6 y 7y3 1
ii 1 0 N y6 __ k) y 1 N 0
Fi)¨
\ NI(4 (:)
\ y4 ,A,0)
r ____________________________________________________
H
Ho OH (BB- 113), O OCH3 (BB- 114),
O 0
NH HNNH
7 Y3 1 /y3 \
y6 ___ II)_yl N S y6 __ () _y1 0
\4 r (:) y4 --1--"0
\ / r __
HO OCH3 (BB- 115), HO F (BB- 116),
O 0
HNNHHNNH
7 \r3 / \1(13 \
y6 y) r
0 Y6 __ P Y1 0
y4 0
\ r __
HO CI (BB- 117), HO OCH3 (BB- 118),
0
0
HNNH /Y3 HNNH
/y3 \
y6 _____ _y1 0 \ y6 __ i r_l
\Y4 /O
r,
()
y4 1-1-1C) \
rL NI :(.4 -1--"0
/r ________________________________________
HO I (BB- 119), Ho CH3(BB- 120),
O 0
HNNH HNNH
/y3 / \r3 \ \
y6 ___ \ i(ii:j4_ yl cr"0 y6 __ F;_y1 r0
y4 0
\ \Y4
r\ _____________________________________________ CH3
r ____________
HO OCH3 (BB- 121), HO OH (BB- 122),
- 56 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0 0
HNNH 7
y /y3 HNNH Y3 \
y6 _______ ilj_l Y6 __ ILY1 "0
Nio H----"Cr
.-
H3C _
HO OCH3 (BB- 123), HO OH (BB- 124), and
0
7y3 HNNH
Y6 ___ ILY1 "0
).õ,õ
.:-
HO 0 (BB- 125), or a
pharmaceutically acceptable salt or stereoisomer thereof,
wherein Y1, Y3, Y4, Y6, and rare as described herein (e.g., each r is,
independently, an integer from 0
to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide is a modified cytidine (e.g., selected from the group
consisting of:
NH2 NH2
y3 \
H3C.,,,.....õ.õ--7-sõõ N
Y3 \ 1
y6 I_y1 I N 0 'I\I
yl
(
y6 _______________________________________ ILi y1 )\ICII
yl 1
0
4 4
Ho OH (BB- 126), Ho OH (BB- 127),
NH2 NH2
/\
HN 'N N
7 y3 7 Y3 \ I
I I N,N0
y6 ___________ ILi y1 y6 __ p y1
yl I
\4 r 0
\Y4
r ___________________________________________________
- __ -
Ho OH (BB- 128), Ho OH (BB- 129),
- 57 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
H3C
NH2 / NH
/
1 N
I /y3 \ 1
v6 v1 N y6 __ ig y1 N 0
\I:
1 I 1
(
y4 10
r _________________________________ I
\Y4
o
.. _
HO OH (BB-130), HO OH (BB-131),
õOH3
NH HN
CH3
/y3\
Y3 \
1 N
y6 __ II:i y1 NO
NI
r _________________________________________ \ N
1
y6 _____________________________________
(4 11:i y1 NO
\!14
-----N: C)
Ho OH (BB- 132), HO OH (BB- 133),
,OH3
HN H3C,N,CH3
\
Y3 \ 1 N
y6 ig y1 N
(
I
AO
r
1 N
I
y6 ___________________________________ \lip/13 y1 N
Y4 0
yI4
r 0
HO oCH3 (BB- 134), Ho OH (BB- 135),
NH2
H3C,N,CH3
HO 1 N
I
(y3 \
\
1N
ig y 1 N
yI
--A- CI \ /y3 \ NO
y6 11:; y1
y6 ___________________________________ I ----Az. )
4
y4 0
Ho OCH3 (BB-136), HO OH ( BB- 137),
- 58 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NHAc NH2
TBDMS
AcON (:)N
1
7y3 \ 1\10 7y3 \ 1\10
v6 __ ig v1 v6 __ P¨ Y1
1 1 1 1 1 1
\ y4 ----I-:. 0
\Y4 ----Az. 0
Ho OH (BB- 138), HO OH (BB- 139),
NH2 NH2
F3C.,,,,.,...õN /N
y3\ 1
ILi v1 N 0
(
y4
y3 \
Y6( 1
Y6( __________________________________ 11:i v1 N 0
I 1
HO 0H (BB- 140), Ho bl-13 (BB- 141),
NH2 NH2
/N \ fN
y3\ y3
1
y6( ILi y1 N ,c,
\!
r ____________ /CH3
y6 ___________________________________ ILi y1 N 0
14
Ho OH (BB- 142), HO 0H (BB- 143),
NH2 NH2
/
7y3 1 N 7Y3 1N
y6 __ ig y1 NO y6 __ ig y1 N
HO 0 (BB- 144), 1-1166(BB- 145),
- 59 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2 NH2
N <N
y3 \ 1
y6 ILi y1 N c;1
( y3
1
y
I ''-i cl
y4 1
y6 ___________________________________ ILi _ N
I '
y4
r *
HO Br (BB- 146), Ho 0H (BB- 147),
NHAc
NH2
N
(y3 \1 N
/N I
1
y6( 11:i y
I '. cl
y4 /y3 \ N 0
_y1 11:ij y1
y4 -----Az. 0
Ho tH3 (BB- 148), HO OH (BB- 149),
NHAc NH2
OHCN
N 1
11:i y1 )
(y3 \ NO
r _____________________ /y3 \ N 0
y6
y6 ___________________________________ II:j v1
\Y4 '
HO oCH3 (BB- 150), HO OH (BB- 151),
NH2 S
OHCN
H3C
1 NN
7 Y3 \ N (:) Y3 \
NH2
y6 __ 11:1 _y1
y6 ______________________________ IlLy1
\4 A 1 0
y4 ----1--
r r
HO OCH3 (BB- 152), Ho OH (BB- 153),
- 60 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
NH2 NH2
BrN Br
/y3 1 /y3 1
II II
y6 _____ p y1 N IC) y6 __ p y1 N IC)
\Y4 C) \(4 C)I \I r r
HO OH (BB- 154), HO OH (BB- 155),
NH2H3C NH2
HON
1 N
1 I
7 \i(13 \ Ncly3 N 0
y6 ___ p y1 y6 ( II:Ly),\, )
\Y4i (4 ---7, v-1 NI
%_, 0
r
.1- --...
HO OH (BB- 156), HO OH (BB- 157),
NH
7 Y3 \ N
IICO2Fmoc
y6 _____ ilj_y1 N NNHFmoc
I
y4 70) H
HO OH (BB- 158), and
NH
7Y3 (NCO2H
y6 _____ ig, ¨y1 NN NH2
I
\Y4 0) H
r _______________
HO OH (BB- 159) , or a pharmaceutically
acceptable
salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and rare as described
herein (e.g., each r is,
independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or
from 1 to 5)). For example,
the building block molecule, which may be incorporated into a polynucleotide
can be:
- 61 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0 0
H 3 C 1 .......... ,.......
1 NH 1 NH
HO-P, -0 NO HO-(17L0 NO
1
\OH zO OH 1r C)
HO OH (BB- 160) or Ho OH (BB-
161), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is,
independently, an integer
from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide is a modified adenosine (e.g., selected from the group
consisting of:
NH2 NH2
(y3 \ y ig 1
N.-..__ NN N
1
r _________________ jCH3 (y3
11
Y6 __ ID yl
1
\Y4
r 0 N
y6
1 )
N----N
/OH
_ __ ..
HO OH (BB- 162), HO tH3 (BB-
163),
NH2 NH2
ig
(Y3
y14
r ,- NN
1
0
----N- y3
y6 __ ig yl
Y14
r ,- N.-..__N
y6 ________ yl N
1
0
N----N-
H3C' ______________________________________________ .
_----._
HO OH (BB- 164), HO 0 (BB-
165),
NH2 NH2
N------N
Y3 \ : 1
y6 11:i yl
(
NI(4 )\1N- Y3 \
Y6 __ ig y1
NI(4 N
1
N-----N-
- 0;-----
HO F (BB- 166), HO CI
(BB- 167),
- 62 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
N H2
N H2
N
N -- --_, --.-.-:-----, I N
Y3 \ N
1 )
( Nr3
y6 _____ II:i yi N --- Nr
NI(4 AO
6
v
'
HO Br (BB- 168), Ho 1
(BB- 169),
N
NH2 H2
(
N,..../1-----___:;N
(
: Yll 3 1 -1
Y3 1 -1
v ________________________________________________________ -----
v6 _____ 11i yl N ----- N
6 p yl N N
' I
' I \ y4 C)
C)
\
\ y4
r ________________________________________________________
r __________________
HO tH3 (BB- 170), Ha OCH3
(BB- 171),
N H2
v6 ig yl N
' YI 3
(
y4
1
so
Ho OH (BB-172),
V 11:1) l N N!\/\F12
' 6 YI 3
(y )
y4
r c 1
HO OH (BB-173),
72
V6 11:1) y1 N ---- N
' Yyl 43 0)
( N---..õ---- -- ---.'=,..HN
Ho OH (BB-174),
- 63 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
NH2
( Y3 \ NN
1
II
y6 __ p y1
N----NOCH3
\4
r ______________
HO OH (BB-175),
NH2
Y3
y6 ig y1
N N S
( N N
1
C---- H3
Nir4 --\,(:)
Nj
HO OH (BB-176),
OH
HN
7Y3 \ NN
1
vo
,
D v1
' ' 1 ' c))\INSCH3
\Y4
r ______________
HO OH (BB-177),
- 64 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
OH
HN
/y3 \
\ 1
y6 __ ig y1
N----NOCH3
\Y4 --0
\ r __
HO OH (BB-178),
OH
HN
/ y 3 \
v6 __ II:i v1 N---N-
1 \\4. 1!17:\.
Ho OH (BB-179),
NH2 NH2
( __
NN Y3 \H C- 1 : r
p- y1 3 N----N v __
Ni4 c, y3
y6 \ \ N
1 A
6 v
ig i N.----N-,
1 I li-A, ,i
y4
/r. \ o 7
Ho OH (BB-180), Ho OH
(BB-181),
NH2 NH2
\ K N
'
Y3 \ ______________ 1 1
I I 1 \ 1
N
( v
o . ----\
II 1 A , N N
y4
/ r \ ON) /y3 \ ________________ N
y6
1 A
y6 ______________________________________ ig y1 N.----N,
\ 1
y4
C)
Ho OH (BB- 182), Ho OH
(BB- 183),
- 65 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
y3 \ F ____________________ r\IN
v ig v------
y4 Y3 \ Br
6 r\IN
v6 _____________________________________ ig y1 N------N
( 1 N N
()
NH: NH:
1 1 Ii'V/ r \ 0, 17 1 y4 1
y3 \ H: or\11-1 IN (BB- 184), Ho
oH (BB- 185),
v6 __ ig v1 N------N
\14 v6 __ ig
NH2
1y3
34 y1\ 1 (:):N:N2
1 Ii'V/ r \ 0, 17 1 1
y3 \ HHosoNH,IN (BB- 186), Ho OH
(BB- 187),
v6 ig v1 N------N
(
y3 \ \S r\IN
v6 ig y1 N------N
y4 o,)
y4
Ci
NH2 NI-1_12
1v 1 I
\i/ r \;(5 OH y1 N.----N
H
v6 ig N (BB- 188), Ho OH (BB-
189),
NH2 NH2
y3 N
/y3 NN1 1\
I ii , 7 1
(
\s _11
y6 _____________________________________ ig
-V0
y4
\ I
y4
C)
Ho OH (BB-190), Ho OH (BB-
191),
- 66 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
\
NH2 NH2
m
Y3 \ S _____________ 1 _iii
ig yl N-----N
(
NI( '-(:; 7Y3 \ ________________ 1 1
y6
II
v6 __ v
D 1 N-----N
4
"I 1
\Y4 0
r ___________________________________________ r ___
Ho OH (BB- 192), HO OH (BB-
193),
NH2 NH2
6 il v
:i 1 N
1 I 1
(
3 H2N NN y6
y4 0 ii N_____Trrsj
v
\ 1
y6 __________________________________ /yP3 y1\ //N CH
\:(4 -VONJ 3
y3 \r _________
6 ig 1
y4 / y3 \
6 _______________________________________ iiii_ N----N
\
Ho cl:N;(BB- 194), y y_1,.H1 b ot)H (BB- 195),
1HN:N HN
(y
;N
v
1 I 1
Ho old (BB- 196), Ho bi-! (BB-
197),
HN
7y3 \ N,õ,------N
1 A
y6 1:_y1 N----N-
I
\Y4 ICI
\ r __ )
Ho OH (BB-198),
- 67 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
HN
( N"----N
y3 1
y6 _____ 11:i yl N N-
y14 C)
r __________________
HO OH (BB- 199), and
HNONH2
N 5
7 Y3 -----N
ii 1
y6 _____ p yl N N-
I
\Y4 C)
\ r __
Ho OH (BB- 200) or a
pharmaceutically
acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and rare as
described herein (e.g.,
each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1
to 3, or from 1 to 5)).
In some embodiments, the building block molecule, which may be incorporated
into a
polynucleotide, is a modified guanosine (e.g., selected from the group
consisting of:
0
Y3
1 NH \
I
N----y ig y1
NNH2
( N
6
(It --()
r _______________________ /CH3
- -
Ho 0H (BB-201),
0
( N
1 NH
Y3 \
I
y6 _____ 11:i yl
N---N-NH2
NI (4 '''\r0
r __________________ /OH
_ ..
_ ..
HO tH3 (BB- 202),
- 68 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
N 0
1 NH
v6 \IID:13 v1 N 1 N NH2
(
y4 , ()
1 ri 1 r ,,
J\JH
Hv6 \IID:13 v N N 2
(
y4 ZNJ
H1r3C: 0)_N N
Ho 01-I (BB- 203),
0
1 Fl 1
y3 \ HO 0 (BB- 204),
N-.....
, 0
( NH
I ,
v6 ______ v1 N ----N-NH2
y4
1 1 1 io
HO F (BB- 205),
0
7y3 \ NH
v6 _____ ig v1
1 I 1 N----NNH2
\Y4 Ao
r
Ho bl (BB- 206),
0
(
y3 \
y6 _____ II:i y1
N-.....
1 NH
N------NNH
Nir4 --0 2
/ r \
HO Br (BB- 207),
- 69 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0
/y3 \ N¨...._
1 NH
v6 v1
\Y4 ANN NH21 I 1
O
r
Ho bl (BB- 208),
0
( NNH
y3 \ 1
vs, ____ ig v1 m----\ . .%\
' \I(4 ' A ,0 j N NH2
/ r \
_ __ /
Ho ol-13 (BB- 209),
0
( zi\I
1 NH
y3 \
\ 1 ,
v6 _____ ig v1 N---N
1 I IssTy
/ r \ (:) NH2
y4
Ha oCH3 (BB-210),
0
( N-,...,/
1 NH
y3
1 ,
v6 _____ ig v1 N----NN
1 I 1
y4 CI H
r __________________
Ho OH (BB-211),
0
( y3 N
NH
y6 _____ 11:1 y1 N---Nrµi
y4 '-V:) H
r __________________
Ho OH (BB-212),
- 70 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
/y3 N1NH
v6 __ ig vi N----NN
1 I 1
\Y4 () H
r ______________
HO 0H (BB-213),
OCH3
7 Y3 \ NN
1
v6 __ ig vl
NI----NNH2
' I 1
\Y4 AO
r
Ho 0H (BB-214),
0
/y3
1
y6 __ II:i yl NI
-----
\ '.i44 AC) NNH, -
\ r __
Ho CI (BB-215),
C)
7Y3 \ 1
y6 __ II:i y1
NI-----NNH
\ '.,4i c4 AO , -
\ r __
Ho old (BB-216),
- 71 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(
Y3
v6 __ 11:i v1
y4 NjC):N
1
oi-----NNH2
1 I 1
r _______________
Ho OH (BB-217),
0
NH
7Y3 \H3C-N 1
v6 __ ig v1 N---NNH2
\y4
Ho 0H (BB-218),
0
/y3 \\ ___________ 1\1...,..NH
v6 __ ig v1
i\INNH2
\Y4
Ho 0H (BB-219),
0
N
/y3 \K(/NH
1
v6 __ ig v1 N---NNH2
1 I 1
Y
\
\ 4 AO
r
Ho OH (BB- 220),
- 72 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0
j\I
( NH
y3 \ ______________ < 1
v6 _____ II:i v1 N---
N ..
1 yl 4 1:{µ _c) N H2
4 V
Ho OH (BB-221),
O
v6 ig v1 N NH2
N
Y3 \ F ______________ r\IC\\IH
(
y4
Ho oi-i (BB- 222),
0
7 Y3 \ CI-1\1NH
Y _____ ig v 1 N---NNH2
1
\y4
Ho oi-i (BB-223),
0
7 Y3 Br __ r\INH
v6 _____ ig v1
1 I 1 N----NNH2
\Y4 0
r _________________
Ho OH (BB- 224),
0
7 y3 \ 1 I\INH
v6 _____ ig v1 N---NNH2
1 I 1
\Y4 AO
r
Ho 01-I (BB- 225),
- 73 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0
7 Y3 \ HS-NH
v6 ________ ig v 1 N---
N NH2
\y4
Ho OH (BB- 226),
0
/y3 \S fNH
v6 ________ ig v 1 N---
N NH2
\y4
HO OH (BB-227),
0
/y3 \ NH
v6 ________ ig v 1 N---
N NH2
\y4
H: ______________ /No I - I (BB-228),
0
y3 \ -...._/.
1 NH
v6 II:i v1 S (N --*
(
1 \1(4 l=-\ ,(:) N NH2
Ho OH (BB- 229),
- 74 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
7 Y3 \ _________ N 1 NH
v6 __ ig v1
NI----NNH2
1 I I'V
\Y4
Ho OH (BB-230),
\
0
( Y3 \ _________ N 1 NH
y6 __ ig y1
NI-----NNH2
, Ao
r
HO OH (BB-231),
0
( N
, NH
y3 1
v6 __ ig v1 N--NN
1 I 1
4 H
y CI
r ______________
Ho OH (BB- 232),
0
( N
, NH
y3 1
v6 __ ig v1 N--NN
1 I 1 1
y4 ()
r ______________
Ho OH (BB-233),
- 75 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Y3 \ N N H
II A
yo ______ p y
NN N H2
NI(4
Ho 0H (BB-234),
Y3 \ NH
y6 ______ p yl
NN N H2
NI(4
Ho 0H (BB-235),
0
Y3 \
y6 ______ p yl
NN N H2
NI(4
Ho 0H (BB- 236), and
0
N N H
/Y3 \ H2N-
y6 ______ p yl
NN N H2
NI(4
Ho 0H (BB- 237), or a pharmaceutically acceptable salt or
stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and rare as described herein
(e.g., each r is,
independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or
from 1 to 5)).
In some embodiments, the chemical modification can include replacement of C
group at
C-5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or
uracil) with N (e.g., replacement
of the >CH group at C-5 with >NRN1 group, wherein RN1 is H or optionally
substituted alkyl). For
example, the building block molecule, which may be incorporated into a
polynucleotide can be:
- 76 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0 0
/\ H3C, -----,..
(
/0 \ HN NH 9 \ N NH
HO _____ I', 0 0 HO
, 1 1
\OH /AO OH /AO
r r
_ _. ...
HO OH (BB- 238) or HO OH (BB- 239) or
0
0
..-----... ..CH3
HN N H3C, ,CH3
/0 \ /0 \ N N
HO ______ P 0
1 Ig 0 0
I
\ OH HO -;--C) \OH AO
_
Ho OH (BB- 240) or HO OH
(BB- 241),
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r
is, independently, an
integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In another embodiment, the chemical modification can include replacement of
the
hydrogen at C-5 of cytosine with halo (e.g., Br, Cl, F, or I) or optionally
substituted alkyl (e.g., methyl).
For example, the building block molecule, which may be incorporated into a
polynucleotide can be:
NH2 NH2
I N H3CN
ii
HO NO ¨0 HO¨¨O N 0
1 1
\OH 2N) Jr--V) \OH ON)
r
HO OH (BB- 242) or HO OH (BB- 243) or
NH2 NHAc
TBDMS,
0, N AcOi N
I 1
0
: Ig, 0
( _______
1
O )
H r _________________ NO 0
OH r :
HO ___________________________________________ P 0 )
1
NO
HO
HO OH (BB- 244) or HO OH
(BB- 245), or a
- 77 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is,
independently, an integer
from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In yet a further embodiment, the chemical modification can include a fused
ring that is
formed by the NH2 at the C-4 position and the carbon atom at the C-5 position.
For example, the
building block molecule, which may be incorporated into a polynucleotide can
be:
H3C
/ NH
N
\
N
HO-P, -0O)O
,
HO OH (BB- 246), or a
pharmaceutically acceptable salt or stereoisomer
thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from
0 to 3, from 1 to 3, or from
1 to 5).
Modifications on the Sugar
The modified nucleosides and nucleotides (e.g., building block molecules),
which may be
incorporated into a polynucleotide (e.g., RNA or mRNA, as described herein),
can be modified on the
sugar of the ribonucleic acid. For example, the 2' hydroxyl group (OH) can be
modified or replaced
with a number of different substituents. Exemplary substitutions at the 2'-
position include, but are not
limited to, H, halo, optionally substituted C1_6 alkyl; optionally substituted
C1_6 alkoxy; optionally
substituted C6_10 aryloxy; optionally substituted C3_5 cycloalkyl; optionally
substituted C3_8 cycloalkoxy;
optionally substituted C6_10 aryloxy; optionally substituted C6_10 aryl-C1_6
alkoxy, optionally substituted
C1_12 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described
herein); a polyethyleneglycol
(PEG), -0(CH2CH20),CH2CH2OR, where R is H or optionally substituted alkyl, and
n is an integer
from 0 to 20 (e.g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from
1 to 4, from 1 to 8, from 1
to 10, from 1 to 16, from 1 to 20, from 2 to 4, from 2 to 8, from 2 to 10,
from 2 to 16, from 2 to 20, from
4 to 8, from 4 to 10, from 4 to 16, and from 4 to 20); "locked" nucleic acids
(LNA) in which the 2'-
hydroxyl is connected by a C1_6 alkylene or C1_6 heteroalkylene bridge to the
4'-carbon of the same
ribose sugar, where exemplary bridges included methylene, propylene, ether, or
amino bridges;
aminoalkyl, as defined herein; aminoalkoxy, as defined herein; amino as
defined herein; and amino
acid, as defined herein
Generally, RNA includes the sugar group ribose, which is a 5-membered ring
having an
oxygen. Exemplary, non-limiting modified nucleotides include replacement of
the oxygen in ribose
(e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a
double bond (e.g., to
replace ribose with cyclopentenyl or cyclohexenyl); ring contraction of ribose
(e.g., to form a 4-
membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to
form a 6- or 7-
membered ring having an additional carbon or heteroatom, such as for
anhydrohexitol, altritol,
- 78 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
mannitol, cyclohexanyl, cyclohexenyl, and morpholino that also has a
phosphoramidate backbone);
multicyclic forms (e.g., tricyclo; and "unlocked" forms, such as glycol
nucleic acid (GNA) (e.g., R-GNA
or S-GNA, where ribose is replaced by glycol units attached to phosphodiester
bonds), threose
nucleic acid (TNA, where ribose is replace with a-L-threofuranosyl-(3'¨>2)) ,
and peptide nucleic acid
(PNA, where 2-amino-ethyl-glycine linkages replace the ribose and
phosphodiester backbone). The
sugar group can also contain one or more carbons that possess the opposite
stereochemical
configuration than that of the corresponding carbon in ribose. Thus, a
polynucleotide molecule can
include nucleotides containing, e.g., arabinose, as the sugar.
Modifications on the Nucleobase
The present disclosure provides for modified nucleosides and nucleotides. As
described
herein "nucleoside" is defined as a compound containing a sugar molecule
(e.g., a pentose or ribose)
or derivative thereof in combination with an organic base (e.g., a purine or
pyrimidine) or a derivative
thereof (also referred to herein as "nucleobase"). As described herein,
"nucleotide" is defined as a
nucleoside including a phosphate group.
Exemplary non-limiting modifications include an amino group, a thiol group, an
alkyl group,
a halo group, or any described herein. The modified nucleotides may by
synthesized by any useful
method, as described herein (e.g., chemically, enzymatically, or recombinantly
to include one or more
modified or non-natural nucleosides).
The modified nucleotide base pairing encompasses not only the standard
adenosine-
thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base
pairs formed between
nucleotides and/or modified nucleotides comprising non-standard or modified
bases, wherein the
arrangement of hydrogen bond donors and hydrogen bond acceptors permits
hydrogen bonding
between a non-standard base and a standard base or between two complementary
non-standard
base structures. One example of such non-standard base pairing is the base
pairing between the
modified nucleotide inosine and adenine, cytosine or uracil.
The modified nucleosides and nucleotides can include a modified nucleobase.
Examples
of nucleobases found in RNA include, but are not limited to, adenine, guanine,
cytosine, and uracil.
Examples of nucleobase found in DNA include, but are not limited to, adenine,
guanine, cytosine, and
thymine. These nucleobases can be modified or wholly replaced to provide
polynucleotide molecules
having enhanced properties, e.g., resistance to nucleases, stability, and
these properties may
manifest through disruption of the binding of a major groove binding partner.
Table 1 below identifies the chemical faces of each canonical nucleotide.
Circles identify
the atoms comprising the respective chemical regions.
- 79 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 1: Chemical faces of each canonical nucleotide.
Watson-Crick
Major Groove Minor Groove Base-pairing
Face Face Face
o 07 crj.Z.`N N
li 7s_r45 - JP 1
Cytidine: 0-P-0 0 N 0
6 0-P-0
OHOH olio
Pyrimidines OH OH
0 0 0
at.... 0
-.1 NH
Uridine: 0-F:70 0 N 0 0-p-O 0 ,tit 0-p707 .... N 0
0 s'
0 0 c?
OHOH OF30H OHOH
NI-12
JR4.0eisl N -, N _O N
Adenosine: Ot7,01sf) N 02FhOW 111150 0-POVI
0 0 6
Purines == . OHOH OHOH
0 0 0
0 1,4 0 H 0N
0 N
<1' I
NR
-71 YI '11
Guanosine: 0-i;.:0-N IsPINH2 0-1-_0 0 1110 2 NI-
f2
0 0 0
OHOH OHOH ol-rm
In some embodiments, B is a modified uracil. Exemplary modified uracils
include those
having Formula (b1)-(b5):
TI\ ;i1" R12c R12c
, R12c
010
N 1 N
V1 NR12a
r2
I I I
Rii Rii.-----õN..------ T N,
c)
' T2 1 T2 1
i (b1), ¨I¨ (b2), I (b3), I (b4), or
0
io
R _Ri2c
N
1
N
(b5), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
is a single or double bond;
each of Tr, T1", Tz, and T2" is, independently, H, optionally substituted
alkyl, optionally
substituted alkoxy, or optionally substituted thioalkoxy, or Tr and T1" or T2'
and T2" join together to
form 0 (oxo), S (thio), or Se (seleno);
each of V1 and V2 is, independently, 0, S, N(Rvb), or C(Rvb), wherein nv is an
integer
from 0 to 2 and each Rvb is, independently, H, halo, optionally substituted
amino acid, optionally
substituted alkyl, optionally substituted haloalkyl, optionally substituted
alkenyl, optionally substituted
alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy,
optionally substituted
- 80 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted
hydroxyalkenyl, optionally
substituted hydroxyalkynyl, optionally substituted aminoalkyl (e.g.,
substituted with an N-protecting
group, such as any described herein, e.g., trifluoroacetyl), optionally
substituted aminoalkenyl,
optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl
(e.g., substituted with an N-
protecting group, such as any described herein, e.g., trifluoroacetyl),
optionally substituted
alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally
substituted
alkoxycarbonylalkynyl, or optionally substituted alkoxycarbonylalkoxy (e.g.,
optionally substituted with
any substituent described herein, such as those selected from (1)-(21) for
alkyl);
R1 is H, halo, optionally substituted amino acid, hydroxyl, optionally
substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted aminoalkyl,
optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl,
optionally substituted
hydroxyalkynyl, optionally substituted aminoalkenyl, optionally substituted
aminoalkynyl, optionally
substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally
substituted
alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl,
optionally substituted
alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally
substituted carboxyalkyl, or
optionally substituted carbamoylalkyl;
R11 is H or optionally substituted alkyl;
R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl,
optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl,
optionally substituted
carboxyalkyl (e.g., optionally substituted with hydroxyl), optionally
substituted carboxyalkoxy,
optionally substituted carboxyaminoalkyl, or optionally substituted
carbamoylalkyl; and
R12 is H, halo, optionally substituted alkyl, optionally substituted alkoxy,
optionally
substituted thioalkoxy, optionally substituted amino, optionally substituted
hydroxyalkyl, optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
Other exemplary modified uracils include those having Formula (b6)-(b9):
Rue Rue
11' T1 R12c
R12a
312a R12b R12a
D12b
,
vv
VV-, VV-Ini2T2 2"
2'
T
(b6), (b7), (b8), or
(b9), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein
is a single or double bond;
each of T1, T1, T2', and T2 is, independently, H, optionally substituted
alkyl, optionally
substituted alkoxy, or optionally substituted thioalkoxy, or T1 and T1 join
together or T2' and T2 join
together (e.g., as in T2) to form 0 (oxo), S (thio), or Se (seleno), or T2 is,
independently, 0 (oxo), S
(thio), or Se (seleno);
each of W1 and W2 is, independently, N(R) nw or C(Rwa),w, wherein nw is an
integer from
0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or
optionally substituted alkoxy;
- 81 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each V3 is, independently, 0, S, N(R), or C(Rva)õ, wherein nv is an integer
from 0 to 2
and each Rva is, independently, H, halo, optionally substituted amino acid,
optionally substituted alkyl,
optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl,
optionally substituted
hydroxyalkynyl, optionally substituted alkenyl, optionally substituted
alkynyl, optionally substituted
heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted
alkoxy, optionally substituted
alkenyloxy, or optionally substituted alkynyloxy, optionally substituted
aminoalkyl (e.g., substituted
with an N-protecting group, such as any described herein, e.g.,
trifluoroacetyl, or sulfoalkyl), optionally
substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally
substituted acylaminoalkyl
(e.g., substituted with an N-protecting group, such as any described herein,
e.g., trifluoroacetyl),
optionally substituted alkoxycarbonylalkyl, optionally substituted
alkoxycarbonylalkenyl, optionally
substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl,
optionally substituted
alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally
substituted with hydroxyl
and/or an 0-protecting group), optionally substituted carboxyalkoxy,
optionally substituted
carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally
substituted with any
substituent described herein, such as those selected from (1)-(21) for alkyl),
and wherein RVa and R12c
taken together with the carbon atoms to which they are attached can form
optionally substituted
cycloalkyl, optionally substituted aryl, or optionally substituted
heterocyclyl (e.g., a 5- or 6-membered
ring);
R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl,
optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, optionally substituted aminoalkynyl,
optionally substituted
carboxyalkyl (e.g., optionally substituted with hydroxyl and/or an 0-
protecting group), optionally
substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl,
optionally substituted
carbamoylalkyl, or absent;
R12b is H, optionally substituted alkyl, optionally substituted alkenyl,
optionally substituted
alkynyl, optionally substituted hydroxyalkyl, optionally substituted
hydroxyalkenyl, optionally
substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally
substituted aminoalkenyl,
optionally substituted aminoalkynyl, optionally substituted alkaryl,
optionally substituted heterocyclyl,
optionally substituted alkheterocyclyl, optionally substituted amino acid,
optionally substituted
alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally
substituted
alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally
substituted
alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally
substituted carboxyalkyl
(e.g., optionally substituted with hydroxyl and/or an 0-protecting group),
optionally substituted
carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally
substituted carbamoylalkyl,
wherein R12b and T1 or R12b and R12 can join together to form optionally
substituted
heterocyclyl; and
R12 is H, halo, optionally substituted alkyl, optionally substituted alkoxy,
optionally
substituted thioalkoxy, optionally substituted amino, optionally substituted
aminoalkyl, optionally
substituted aminoalkenyl, or optionally substituted aminoalkynyl.
Further exemplary modified uracils include those having Formula (b28)-(b31):
- 82 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
T1 T1 T1
Vb' Vb'
R N R1 2a
R N R1 2a
R12 R1 2a
N N
Rvb"/\
" 2TT2
I
(b28), (b29), (b30), or
T1
RVb' N 12a
NT2
(b31), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
each of Tland T2 is, independently, 0 (oxo), S (thio), or Se (seleno);
each Rvb and Rvb is, independently, H, halo, optionally substituted amino
acid, optionally
substituted alkyl, optionally substituted haloalkyl, optionally substituted
hydroxyalkyl, optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted alkenyl,
optionally substituted alkynyl, optionally substituted alkoxy, optionally
substituted alkenyloxy,
optionally substituted alkynyloxy, optionally substituted aminoalkyl (e.g.,
substituted with an N-
protecting group, such as any described herein, e.g., trifluoroacetyl, or
sulfoalkyl), optionally
substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally
substituted acylaminoalkyl
(e.g., substituted with an N-protecting group, such as any described herein,
e.g., trifluoroacetyl),
optionally substituted alkoxycarbonylalkyl, optionally substituted
alkoxycarbonylalkenyl, optionally
substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl,
optionally substituted
alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally
substituted with hydroxyl
and/or an 0-protecting group), optionally substituted carboxyalkoxy,
optionally substituted
carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally
substituted with any
substituent described herein, such as those selected from (1)-(21) for alkyl)
(e.g., Rvb is optionally
substituted alkyl, optionally substituted alkenyl, or optionally substituted
aminoalkyl, e.g., substituted
with an N-protecting group, such as any described herein, e.g.,
trifluoroacetyl, or sulfoalkyl);
2012a =
R
H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl,
optionally
substituted aminoalkyl (e.g., e.g., substituted with an N-protecting group,
such as any described
herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted
aminoalkenyl, or optionally substituted
aminoalkynyl; and
R12b is H, optionally substituted hydroxyl, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl,
optionally substituted
hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted
aminoalkyl, optionally
substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g.,
substituted with an N-protecting
group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl),
optionally substituted
alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally
substituted
alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally
substituted
alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally
substituted
carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted
carbamoylalkyl.
- 83 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In particular embodiments, T1 is 0 (oxo), and T2 is S (thio) or Se (seleno).
In other
embodiments, T1 is S (thio), and T2 is 0 (oxo) or Se (seleno). In some
embodiments, Rvb is H,
optionally substituted alkyl, or optionally substituted alkoxy.
In other embodiments, each R12a and R12b is, independently, H, optionally
substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, or optionally
substituted hydroxyalkyl. In
particular embodiments, R12a is H. In other embodiments, both R12a and R12b
are H.
In some embodiments, each Rvb and R12b is, independently, optionally
substituted
aminoalkyl (e.g., substituted with an N-protecting group, such as any
described herein, e.g.,
trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl,
optionally substituted aminoalkynyl,
or optionally substituted acylaminoalkyl (e.g., substituted with an N-
protecting group, such as any
described herein, e.g., trifluoroacetyl). In some embodiments, the amino
and/or alkyl of the optionally
substituted aminoalkyl is substituted with one or more of optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted
carboxy (e.g., substituted
with an 0-protecting group), optionally substituted hydroxyl (e.g.,
substituted with an 0-protecting
group), optionally substituted carboxyalkyl (e.g., substituted with an 0-
protecting group), optionally
substituted alkoxycarbonylalkyl (e.g., substituted with an 0-protecting
group), or N-protecting group.
In some embodiments, optionally substituted aminoalkyl is substituted with an
optionally substituted
sulfoalkyl or optionally substituted alkenyl. In particular embodiments, R12a
and Rvb are both H. In
particular embodiments, T1 is 0 (oxo), and T2 is S (thio) or Se (seleno).
In some embodiments, Rvb is optionally substituted alkoxycarbonylalkyl or
optionally
substituted carbamoylalkyl.
In particular embodiments, the optional substituent for R12a5 R121D5 R12c, or
RVa is a
polyethylene glycol group (e.g., -(CH2)s2(OCH2C1-12)s1(C1-12)s3OR', wherein 51
is an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an
integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R'
is H or C1_20 alkyl); or an
amino-polyethylene glycol group (e.g., -NRN1 (CH2)s2(CH2CH20)si (C1-12)s3NRN1,
wherein 51 is an
integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer
from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or
from 1 to 10), and each RN1 is,
independently, hydrogen or optionally substituted C1_6 alkyl).
In some embodiments, B is a modified cytosine. Exemplary modified cytosines
include
compounds of Formula (b10)-(b14):
D 13a NR13b ,R13b R13a R13b R13a
R13b
,
IN
14
R14v5 R ,R16
N N V4

15 /\
R15N\---3T3 R N 3.3
R15
T T T T3'
(131 0), (b11), (b12),
(b13),
\/4N
R15 -r3÷
Ta
or (b14), or a pharmaceutically acceptable salt or
stereoisomer thereof, wherein
- 84 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each T3" is, independently, H, optionally substituted alkyl, optionally
substituted alkoxy, or
optionally substituted thioalkoxy, or T3' and T3" join together (e.g., as in
T3) to form 0 (oxo), S (thio), or
Se (seleno);
each V4 is, independently, 0, S, N(Rvc)õ, or C(Rvc)õ, wherein nv is an integer
from 0 to 2
and each Rvc is, independently, H, halo, optionally substituted amino acid,
optionally substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted alkoxy, optionally
substituted alkenyloxy, optionally substituted heterocyclyl, optionally
substituted alkheterocyclyl, or
optionally substituted alkynyloxy (e.g., optionally substituted with any
substituent described herein,
such as those selected from (1)-(21) for alkyl);
each V5 is, independently, N(Rvd)õ, or C(Rvd)õ, wherein nv is an integer from
0 to 1 and
each Rvd is, independently, H, halo, optionally substituted amino acid,
optionally substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted alkoxy, optionally
substituted alkenyloxy, optionally substituted heterocyclyl, optionally
substituted alkheterocyclyl, or
optionally substituted alkynyloxy (e.g., optionally substituted with any
substituent described herein,
such as those selected from (1)-(21) for alkyl) (e.g., V5 is ¨CH or N);
each of R13a and R13b is, independently, H, optionally substituted acyl,
optionally
substituted acyloxyalkyl, optionally substituted alkyl, or optionally
substituted alkoxy, wherein R13b and
R14 can be taken together to form optionally substituted heterocyclyl;
each R14 is, independently, H, halo, hydroxyl, thiol, optionally substituted
acyl, optionally
substituted amino acid, optionally substituted alkyl, optionally substituted
haloalkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
hydroxyalkyl (e.g., substituted
with an 0-protecting group), optionally substituted hydroxyalkenyl, optionally
substituted
hydroxyalkynyl, optionally substituted alkoxy, optionally substituted
alkenyloxy, optionally substituted
alkynyloxy, optionally substituted aminoalkoxy, optionally substituted
alkoxyalkoxy, optionally
substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R
is H, alkyl, aryl, or
phosphoryl), azido, optionally substituted aryl, optionally substituted
heterocyclyl, optionally
substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally
substituted aminoalkenyl, or
optionally substituted aminoalkynyl; and
each of R15 and R16 is, independently, H, optionally substituted alkyl,
optionally substituted
alkenyl, or optionally substituted alkynyl.
Further exemplary modified cytosines include those having Formula (b32)-(b35):
R13a ,R13b N,R13b T1 R13aN,R13b
014R14 16 R14õ,
N R14
15 313a
R R
NT3 N
, 13b
(b32), ,
(b33), (b34), or I
(b35),
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
each of T1 and T3 is, independently, 0 (oxo), S (thio), or Se (seleno);
- 85 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
each of R13a and R13b is, independently, H, optionally substituted acyl,
optionally
substituted acyloxyalkyl, optionally substituted alkyl, or optionally
substituted alkoxy, wherein R13b and
R14 can be taken together to form optionally substituted heterocyclyl;
each R14 is, independently, H, halo, hydroxyl, thiol, optionally substituted
acyl, optionally
substituted amino acid, optionally substituted alkyl, optionally substituted
haloalkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
hydroxyalkyl (e.g., substituted
with an 0-protecting group), optionally substituted hydroxyalkenyl, optionally
substituted
hydroxyalkynyl, optionally substituted alkoxy, optionally substituted
alkenyloxy, optionally substituted
alkynyloxy, optionally substituted aminoalkoxy, optionally substituted
alkoxyalkoxy, optionally
substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R
is H, alkyl, aryl, or
phosphoryl), azido, optionally substituted aryl, optionally substituted
cycloalkyl, optionally substituted
heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted
aminoalkyl (e.g.,
hydroxyalkyl, alkyl, alkenyl, or alkynyl), optionally substituted
aminoalkenyl, or optionally substituted
aminoalkynyl; and
each of R15 and R16 is, independently, H, optionally substituted alkyl,
optionally substituted
alkenyl, or optionally substituted alkynyl (e.g., R15 is H, and R16 is H or
optionally substituted alkyl).
In some embodiments, R15 is H, and R16 is H or optionally substituted alkyl.
In particular
embodiments, R14 is H, acyl, or hydroxyalkyl. In some embodiments, R14 is
halo. In some
embodiments, both R14 and R15 are H. In some embodiments, both R15 and R16 are
H. In some
embodiments, each of R14 and R15 and R16 is H. In further embodiments, each of
R13a and R13b is
independently, H or optionally substituted alkyl.
Further non-limiting examples of modified cytosines include compounds of
Formula (b36):
,R13b
14a
N
R15 N R1 4b
(b36) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
each R13b is, independently, H, optionally substituted acyl, optionally
substituted
acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy,
wherein R13b and R14b can
be taken together to form optionally substituted heterocyclyl;
each R14a and R14b is, independently, H, halo, hydroxyl, thiol, optionally
substituted acyl,
optionally substituted amino acid, optionally substituted alkyl, optionally
substituted haloalkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted hydroxyalkyl (e.g.,
substituted with an 0-protecting group), optionally substituted
hydroxyalkenyl, optionally substituted
alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy,
optionally substituted
aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted
acyloxyalkyl, optionally
substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, phosphoryl,
optionally substituted
aminoalkyl, or optionally substituted carboxyaminoalkyl), azido, optionally
substituted aryl, optionally
substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally
substituted aminoalkyl,
optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl;
and
- 86 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of R15 is, independently, H, optionally substituted alkyl, optionally
substituted alkenyl,
or optionally substituted alkynyl.
In particular embodiments, R14b is an optionally substituted amino acid (e.g.,
optionally
substituted lysine). In some embodiments, R14a is H.
In some embodiments, B is a modified guanine. Exemplary modified guanines
include
compounds of Formula (b15)-(b17):
T4' T4" T5' \ T5" R23' "
T5 05
vc ,R18 V
N N-_____\ Nõ---- N------N,R18
V R21 \ __ R24 R17 1
\
NN 019a T6"
N------F` 1 N
N------:1-------N N----N\-rb' 1
1 R19b
(b15), 1 R22
(b16), or ¨1¨ R22
(b17),
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
Each of T4, T4, T5', T5, T6, and T6 is, independently, H, optionally
substituted alkyl, or
optionally substituted alkoxy, and wherein T4 and T4 (e.g., as in T4) or T5'
and T5 or T6 and T6 join
together to form 0 (oxo), S (thio), or Se (seleno);
each of V5 and V6 is, independently, 0, S, N(Rvd)õ, or C(Rvd)õ, wherein nv is
an integer
from 0 to 2 and each Rvd is, independently, H, halo, thiol, optionally
substituted amino acid, cyano,
amidine, optionally substituted aminoalkyl, optionally substituted
aminoalkenyl, optionally substituted
aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl,
optionally substituted alkynyl,
optionally substituted alkoxy, optionally substituted alkenyloxy, optionally
substituted alkynyloxy (e.g.,
optionally substituted with any substituent described herein, such as those
selected from (1)-(21) for
alkyl), optionally substituted thioalkoxy, or optionally substituted amino;
and
each of R17, R185 R19a5 R19b5 R215 R225 1-K.¨.235
and R24 is, independently, H, halo, thiol,
optionally substituted alkyl, optionally substituted alkenyl, optionally
substituted alkynyl, optionally
substituted thioalkoxy, optionally substituted amino, or optionally
substituted amino acid.
Exemplary modified guanosines include compounds of Formula (b37)-(b40):
T4 T4' T4
NN-R18
N _
// ------1' N
\ 019a / N R18
019a
1
N N NR
1 1 1
, R19b
(b37), ' R19b
(b38), ' R19b
(b39), or
T4
N__N- R18
R21 _______ </
N----NN,R19a
I I
, R19b
(b40), or a pharmaceutically acceptable salt or stereoisomer thereof,
wherein
each of -14 is, independently, H, optionally substituted alkyl, or optionally
substituted
alkoxy, and each T4 is, independently, 0 (oxo), S (thio), or Se (seleno);
- 87 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
each of R18, R19a, R19b, and R21 is, independently, H, halo, thiol, optionally
substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted thioalkoxy,
optionally substituted amino, or optionally substituted amino acid.
In some embodiments, R18 is H or optionally substituted alkyl. In further
embodiments, T4
is oxo. In some embodiments, each of R19a and R19b is, independently, H or
optionally substituted
alkyl.
In some embodiments, B is a modified adenine. Exemplary modified adenines
include
compounds of Formula (b18)-(b20):
R26a R26 b
R26b
R29
N 7 ,-,28 V7 N
R25 ________________________ R25 ___________________ R25 __
R27 R27
(b18), (b19), or
(b20),
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
each V7 is, independently, 0, S, N(R), or C(Rve)õ, wherein nv is an integer
from 0 to 2
and each Rve is, independently, H, halo, optionally substituted amino acid,
optionally substituted alkyl,
optionally substituted alkenyl, optionally substituted alkynyl, optionally
substituted alkoxy, optionally
substituted alkenyloxy, or optionally substituted alkynyloxy (e.g., optionally
substituted with any
substituent described herein, such as those selected from (1)-(21) for alkyl);
each R25 is, independently, H, halo, thiol, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or
optionally substituted
amino;
each of R26a and R26b is, independently, H, optionally substituted acyl,
optionally
substituted amino acid, optionally substituted carbamoylalkyl, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
hydroxyalkyl, optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted alkoxy, or
polyethylene glycol group (e.g., -(C1-12)s2(OCH2C1-12)s1 (CI-12)s3OR', wherein
s1 is an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an
integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R'
is H or C1_20 alkyl); or an
amino-polyethylene glycol group (e.g., -NRN1 (C1-12)s2(CH2CH20)si (C1-
12)s3NRN1, wherein s1 is an
integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer
from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or
from 1 to 10), and each RN1 is,
independently, hydrogen or optionally substituted C1_6 alkyl);
each R27 is, independently, H, optionally substituted alkyl, optionally
substituted alkenyl,
optionally substituted alkynyl, optionally substituted alkoxy, optionally
substituted thioalkoxy, or
optionally substituted amino;
each R28 is, independently, H, optionally substituted alkyl, optionally
substituted alkenyl, or
optionally substituted alkynyl; and
each R29 is, independently, H, optionally substituted acyl, optionally
substituted amino
acid, optionally substituted carbamoylalkyl, optionally substituted alkyl,
optionally substituted alkenyl,
- 88 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
optionally substituted alkynyl, optionally substituted hydroxyalkyl,
optionally substituted
hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted
amino.
Exemplary modified adenines include compounds of Formula (b41)-(b43):
R26a R26b R26a R26b R26a R26b
N
R25 _________________________
(b41), (b42), or -1- (b43), or a
pharmaceutically acceptable salt or stereoisomer thereof, wherein
each R25 is, independently, H, halo, thiol, optionally substituted alkyl,
optionally substituted
alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or
optionally substituted
amino;
each of R26a and R26b is, independently, H, optionally substituted acyl,
optionally
substituted amino acid, optionally substituted carbamoylalkyl, optionally
substituted alkyl, optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
hydroxyalkyl, optionally
substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally
substituted alkoxy, or
polyethylene glycol group (e.g., -(C1-12)s2(OCH2C1-12)s1 (CI-12)s3OR', wherein
51 is an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an
integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R'
is H or C1_20 alkyl); or an
amino-polyethylene glycol group (e.g., -NRN1 (C1-12)s2(CH2CH20)si (C1-
12)s3NRN1, wherein 51 is an
integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer
from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or
from 1 to 10), and each RN1 is,
independently, hydrogen or optionally substituted C1_6 alkyl); and
each R27 is, independently, H, optionally substituted alkyl, optionally
substituted alkenyl,
optionally substituted alkynyl, optionally substituted alkoxy, optionally
substituted thioalkoxy, or
optionally substituted amino.
In some embodiments, R26a is H, and R26b is optionally substituted alkyl. In
some
embodiments, each of R26a and R26b is, independently, optionally substituted
alkyl. In particular
embodiments, R27 is optionally substituted alkyl, optionally substituted
alkoxy, or optionally substituted
thioalkoxy. In other embodiments, R25 is optionally substituted alkyl,
optionally substituted alkoxy, or
optionally substituted thioalkoxy.
In particular embodiments, the optional substituent for R26a, R26b, or R29 is
a polyethylene
glycol group (e.g., -(C1-12)s2(OCH2C1-12)s1 (C1-12)s3OR', wherein 51 is an
integer from 1 to 10 (e.g., from 1
to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0
to 10 (e.g., from 0 to 4,
from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or C1_20
alkyl); or an amino-
polyethylene glycol group (e.g., -NRN1 (C1-12)s2(CH2CH20)si (C1-12)s3NRN1,
wherein s1 is an integer from
1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently,
is an integer from 0 to 10
(e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10),
and each RN1 is,
independently, hydrogen or optionally substituted C1_6 alkyl).
- 89 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
In some embodiments, B may have Formula (b21):
ex12
1 2 a
FX
T2
(b21), wherein X12 is, independently, 0, S, optionally substituted alkylene
(e.g.,
methylene), or optionally substituted heteroalkylene, xa is an integer from 0
to 3, and R12a and T2 are
as described herein.
In some embodiments, B may have Formula (b22):
0 T1
R10'
,R12a
N N
R11 2
N T2
(b22), wherein R1 ' is, independently, optionally substituted alkyl,
optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
aryl, optionally substituted
heterocyclyl, optionally substituted aminoalkyl, optionally substituted
aminoalkenyl, optionally
substituted aminoalkynyl, optionally substituted alkoxy, optionally
substituted alkoxycarbonylalkyl,
optionally substituted alkoxycarbonylalkenyl, optionally substituted
alkoxycarbonylalkynyl, optionally
substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy,
optionally substituted
carboxyalkyl, or optionally substituted carbamoylalkyl, and R11, R12a, 1-15
and T2 are as described
herein.
In some embodiments, B may have Formula (b23):
Rio N.R12a
R T2
(b23), wherein R1 is optionally substituted heterocyclyl (e.g., optionally
substituted fury!, optionally substituted thienyl, or optionally substituted
pyrrolyl), optionally substituted
aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl),
or any substituent
described herein (e.g., for R10) ;and wherein R11 (e.g., H or any substituent
described herein), R12a
(e.g., H or any substituent described herein), T1 (e.g., oxo or any
substituent described herein), and
T2 (e.g., oxo or any substituent described herein) are as described herein.
In some embodiments, B may have Formula (b24):
R13a , R13b
0
R14' n
R15 N'-r3
(b24), wherein R14' is, independently, optionally substituted alkyl,
optionally
substituted alkenyl, optionally substituted alkynyl, optionally substituted
aryl, optionally substituted
- 90 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
heterocyclyl, optionally substituted alkaryl, optionally substituted
alkheterocyclyl, optionally substituted
aminoalkyl, optionally substituted aminoalkenyl, optionally substituted
aminoalkynyl, optionally
substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally
substituted
alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl,
optionally substituted
alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally
substituted carboxyalkyl, or
optionally substituted carbamoylalkyl, and R13a, R13b, K-15,
and T3 are as described herein.
In some embodiments, B may have Formula (b25):
3a D13b
0
R15
(b25), wherein R14 is optionally substituted heterocyclyl (e.g., optionally
substituted fury!, optionally substituted thienyl, or optionally substituted
pyrrolyl), optionally substituted
aryl (e.g., optionally substituted phenyl or optionally substituted naphthyl),
or any substituent
described herein (e.g., for R14 or R14); and wherein R13a (e.g., H or any
substituent described herein),
Ri3b (e.g., H or any substituent described herein), R15 (e.g., H or any
substituent described herein),
and T3 (e.g., oxo or any substituent described herein) are as described
herein.
In some embodiments, B is a nucleobase selected from the group consisting of
cytosine,
guanine, adenine, and uracil. In some embodiments, B may be:
N-N7 NH2 0
N
(b26) or (b27).
In some embodiments, the modified nucleobase is a modified uracil. Exemplary
nucleobases and nucleosides having a modified uracil include pseudouridine
(t.p), pyridin-4-one
ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2-thio-
uridine (s2U), 4-thio-uridine
(s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-
aminoallyl-uridine, 5-
halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 3-methyl-uridine
(m3U), 5-methoxy-uridine
(mo5U), uridine 5-oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl
ester (mcmo5U), 5-
carboxymethyl-uridine (cm5U), 1-carboxymethyl-pseudouridine, 5-
carboxyhydroxymethyl-uridine
(chm5U), 5-carboxyhydroxymethyl-uridine methyl ester (mchm5U), 5-
methoxycarbonylmethyl-uridine
(mcm5U), 5-methoxycarbonylmethy1-2-thio-uridine (mcm5S2U), 5-aminomethy1-2-
thio-uridine (nm5S2U),
5-methylaminomethyl-uridine (mnm5U), 5-methylaminomethy1-2-thio-uridine
(mnm5s2U), 5-
methylaminomethy1-2-seleno-uridine (mnm5se2U), 5-carbamoylmethyl-uridine
(ncm5U), 5-
carboxymethylaminomethyl-uridine (cmnm5U), 5-carboxymethylaminomethy1-2-thio-
uridine
(cmnm5S2U), 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyl-
uridine (tm5U), 1-
taurinomethyl-pseudouridine, 5-taurinomethy1-2-thio-uridine(Tm5s2U), 1-
taurinomethy1-4-thio-
pseudouridine, 5-methyl-uridine (m5U, i.e., having the nucleobase
deoxythymine), 1-methyl-
pseudouridine
5-methyl-2-thio-uridine (m5S2U), 1-methyl-4-thio-pseudouridine (m1S4t.p), 4-
thio-
1-methyl-pseudouridine, 3-methyl-pseudouridine (m3t.p), 2-thio-1-methyl-
pseudouridine, 1-methy1-1-
- 91 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
deaza-pseudouridine, 2-thio-1-methy1-1-deaza-pseudouridine, dihydrouridine
(D),
dihydropseudouridine, 5,6-dihydrouridine, 5-methyl-dihydrouridine (m5D), 2-
thio-dihydrouridine, 2-
thio-dihydropseudouridine, 2-methoxy-uridine, 2-methoxy-4-thio-uridine, 4-
methoxy-pseudouridine, 4-
methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 3-(3-amino-3-
carboxypropyl)uridine
(acp3U), 1-methy1-3-(3-amino-3-carboxypropyl)pseudouridine (acp3 t.p), 5-
(isopentenylaminomethyl)uridine (inm5U), 5-(isopentenylaminomethyl)-2-thio-
uridine (inm5s2U), a-thio-
uridine, 2'-0-methyl-uridine (Urn), 5,2'-0-dimethyl-uridine (m5Um), 2'-0-
methyl-pseudouridine (t.pm),
2-thio-2'-0-methyl-uridine (s2Um), 5-methoxycarbonylmethy1-2'-0-methyl-uridine
(mcm5Um), 5-
carbamoylmethy1-2'-0-methyl-uridine (ncm5Um), 5-carboxymethylaminomethy1-2'-0-
methyl-uridine
(cmnm5Um), 3,2'-0-dimethyl-uridine (m3Um), 5-(isopentenylaminomethyl)-2'-0-
methyl-uridine
(inm5Um), 1-thio-uridine, deoxythymidine, 2'-F-ara-uridine, 2'-F-uridine, 2'-
OH-ara-uridine,
5-(2-carbomethoxyvinyl) uridine, and 543-(1-E-propenylamino)uridine.
In some embodiments, the modified nucleobase is a modified cytosine. Exemplary
nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine,
6-aza-cytidine,
pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (act), 5-formyl-
cytidine (f5C), N4-
methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo-cytidine (e.g., 5-iodo-
cytidine), 5-
hydroxymethyl-cytidine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo-cytidine,
pyrrolo-
pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio-
pseudoisocytidine, 4-thio-1-
methyl-pseudoisocytidine, 4-thio-1-methy1-1-deaza-pseudoisocytidine, 1-methy1-
1-deaza-
pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-
thio-zebularine, 2-thio-
zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-
pseudoisocytidine, 4-
methoxy-1-methyl-pseudoisocytidine, lysidine (k2C), a-thio-cytidine, 2'-0-
methyl-cytidine (Cm), 5,2'-0-
dimethyl-cytidine (m5Cm), N4-acetyl-2'-0-methyl-cytidine (ac4Cm), N4,2'-0-
dimethyl-cytidine (m4Cm),
5-formy1-2'-0-methyl-cytidine (f5Cm), N4,N4,2'-0-trimethyl-cytidine (m42Cm), 1-
thio-cytidine,
2'-F-ara-cytidine, 2'-F-cytidine, and 2'-OH-ara-cytidine.
In some embodiments, the modified nucleobase is a modified adenine. Exemplary
nucleobases and nucleosides having a modified adenine include 2-amino-purine,
2, 6-diaminopurine,
2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-
chloro-purine), 2-amino-
6-methyl-purine, 8-azido-adenosine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-
deaza-2-amino-
purine, 7-deaza-8-aza-2-amino-purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-
2,6-diaminopurine,
1-methyl-adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl-adenosine (m6A), 2-
methylthio-N6-
methyl-adenosine (ms2m6A), N6-isopentenyl-adenosine (i6A), 2-methylthio-N6-
isopentenyl-adenosine
2.
(MS I6 A), N6-(cis-hydroxyisopentenyl)adenosine (io6A), 2-methylthio-N6-(cis-
hydroxyisopentenyl)adenosine (ms2io6 A), N6-glycinylcarbamoyl-adenosine (g6A),
N6-
threonylcarbamoyl-adenosine (t6A), N6-methyl-N6-threonylcarbamoyl-adenosine
(m6t6A), 2-
methylthio-N6-threonylcarbamoyl-adenosine (ms2g6A), N6,N6-dimethyl-adenosine
(m62A), N6-
hydroxynorvalylcarbamoyl-adenosine (hn6A), 2-methylthio-N6-
hydroxynorvalylcarbamoyl-adenosine
(ms2hn6A), N6-acetyl-adenosine (ac6A), 7-methyl-adenine, 2-methylthio-adenine,
2-methoxy-adenine,
a-thio-adenosine, 2'-0-methyl-adenosine (Am), N6,2'-0-dimethyl-adenosine
(m6Am), N6, N6,2'-O-
trimethyl-adenosine (m62Am), 1,2'-0-dimethyl-adenosine (mlAm), 2'-0-
ribosyladenosine (phosphate)
- 92 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(Ar(p)), 2-amino-N6-methyl-purine, 1-thio-adenosine, 8-azido-adenosine, 2'-F-
ara-adenosine,
2'-F-adenosine, 2'-0H-ara-adenosine, and N6-(19-amino-pentaoxanonadecyI)-
adenosine.
In some embodiments, the modified nucleobase is a modified guanine. Exemplary
nucleobases and nucleosides having a modified guanine include inosine (I), 1-
methyl-inosine (m11),
wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG-14), isowyosine
(imG2),
wybutosine (yVV), peroxywybutosine (o2yVV), hydroxywybutosine (OhyVV),
undermodified
hydroxywybutosine (OhyW*), 7-deaza-guanosine, queuosine (Q), epoxyqueuosine
(oQ), galactosyl-
queuosine (galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine
(preQ0), 7-
aminomethy1-7-deaza-guanosine (preQi), archaeosine (G ), 7-deaza-8-aza-
guanosine, 6-thio-
guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-
guanosine (m7G),
6-thio-7-methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-
guanosine (m1G), N2-
methyl-guanosine (m2G), N2,N2-dimethyl-guanosine (m22G), N2,7-dimethyl-
guanosine (m27G), N2,
N2,7-dimethyl-guanosine (m227G), 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-
methy1-6-thio-
guanosine, N2-methyl-6-thio-guanosine, N2,N2-dimethy1-6-thio-guanosine, a-thio-
guanosine, 2'-0-
methyl-guanosine (Gm), N2-methyl-2'-0-methyl-guanosine (m2Gm), N2,N2-dimethy1-
2'-0-methyl-
guanosine (m22Gm), 1-methy1-2'-0-methyl-guanosine (m1Gm), N2,7-dimethy1-2'-0-
methyl-guanosine
(m2 7Gm), 2'-0-methyl-inosine (Im), 1,2'-0-dimethyl-inosine (mllm), 2'-0-
ribosylguanosine
(phosphate) (Gr(p)) , 1-thio-guanosine, 06-methyl-guanosine, 2'-F-ara-
guanosine, and
2'-F-guanosine.
In some embodiments, the nucleotide can be modified. For example, such
modifications
include replacing hydrogen on C-5 of uracil or cytosine with alkyl (e.g.,
methyl) or halo.
The nucleobase of the nucleotide can be independently selected from a purine,
a
pyrimidine, a purine or pyrimidine analog. For example, the nucleobase can
each be independently
selected from adenine, cytosine, guanine, uracil, or hypoxanthine. In another
embodiment, the
nucleobase can also include, for example, naturally-occurring and synthetic
derivatives of a base,
including pyrazolo[3,4-d]pyrimidines, 5-methylcytosine (5-me-C), 5-
hydroxymethyl cytosine, xanthine,
hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine
and guanine, 2-propyl
and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-
thiothymine and 2-thiocytosine, 5-
propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil
(pseudouracil), 4-thiouracil,
8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-
substituted adenines and
guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-
substituted uracils and cytosines,
7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine,
deazaguanine, 7-
deazaguanine, 3-deazaguanine, deazaadenine, 7-deazaadenine, 3-deazaadenine,
pyrazolo[3,4-
d]pyrimidine, imidazo[1,5-a]1,3,5 triazinones, 9-deazapurines, imidazo[4,5-
d]pyrazines, thiazolo[4,5-
d]pyrimidines, pyrazin-2-ones, 1,2,4-triazine, pyridazine; and 1,3,5 triazine.
When the nucleotides are
depicted using the shorthand A, G, C, T or U, each letter refers to the
representative base and/or
derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-
deaza adenine).
- 93 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
In some embodiments, the modified nucleotide is a compound of Formula XI:
p_ B
ORcl
n \ /Z _
D A
XI
wherein:
denotes a single or a double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or ¨CRaRb- when
denotes a single bond, or U is ¨CRa- when
denotes a double bond;
Z is H, C1_12 alkyl, or C6_20 aryl, or Z is absent when
denotes a double bond; or
Z is ¨CRaRb- and forms a bond with A;
A is H, OH, NHR wherein R= alkyl or aryl or phosphoryl, sulfate, -NH2, N3,
azido, -SH, an
amino acid, or a peptide comprising 1 to 12 amino acids;
D is H, OH, NHR wherein R= alkyl or aryl or phosphoryl, -NH2, -SH, an amino
acid, a
peptide comprising 1 to 12 amino acids, or a group of Formula XII:
(y2\
Yl ______________ =x
ORcli
XII
or A and D together with the carbon atoms to which they are attached form a 5-
membered
ring;
X is 0 or S;
each of Y1 is independently selected from _oRa1, _NRa1RID1, and ¨SRal;
each of Y2 and Y3 are independently selected from 0, -CRaRb-, NRc, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
n is 0, 1,2, or 3;
m is 0, 1, 2 or 3;
B is nucleobase;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai and Rbi are each independently H or a counterion; and
_OR cl is OH at a pH of about 1 or ¨ORcl is 0- at physiological pH;
provided that the ring encompassing the variables A, B, D, U, Z, Y2 and Y3
cannot be
ribose.
- 94 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, B is a nucleobase selected from the group consisting of
cytosine,
guanine, adenine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In some embodiments, the modified nucleotides are a compound of Formula Xl-a:
/X \
Y1 _______________ y2y3 B
ORcl
n\
0\ /0
Xl-a.
In some embodiments, the modified nucleotides are a compound of Formula Xl-b:
/X \
_______________ 11:;_y_ay3 B
ORcl
n \
HO OH
Xl-b.
In some embodiments, the modified nucleotides are a compound of Formula Xl-c1,
Xl-c2,
or Xl-c3:
/X \ /X \ /X \
Y1 _____ p y_ay3 B c) y2y3 B I _______ y3
'
cl /X ORcl
ORcl *
n\
n\OR n \
HO A HO OH HO A
Xl-c1 Xl-c2 Xl-c3
In some embodiments, the modified nucleotides are a compound of Formula XI:
(X
Y1 _____________ ig
0 Rci U
n \
D A
XI
wherein:
denotes a single or a double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or ¨CRaRb- when denotes a single bond, or U is ¨CRa- when
denotes a double bond;
Z is H, C1_12 alkyl, or C6_20 aryl, or Z is absent when denotes a double
bond; or
Z is ¨CRaRb- and forms a bond with A;
- 95 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
A is H, OH, sulfate, -NH2, -SH, an amino acid, or a peptide comprising 1 to 12
amino
acids;
D is H, OH, -NH2, -SH, an amino acid, a peptide comprising 1 to 12 amino
acids, or a
group of Formula XII:
/
/
(al-
y2
yl ____________ il=x
1
ORcim
XII
or A and D together with the carbon atoms to which they are attached form a 5-
membered
ring;
X is 0 or S;
each of Y1 is independently selected from _oRai, _NRalRbl, and ¨SRai ;
each of Y2 and Y3 are independently selected from 0, -CRaRb-, NRb, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
n is 0, 1,2, or 3;
m is 0, 1, 2 or 3;
B is a nucleobase of Formula XIII:
R3
V.¨.,.....---1-----...-N
R44 1
N----*-Nr"---R5
XIII
wherein:
V is N or positively charged NW;
R3 is NWRd, -0Ra, or ¨SRa;
R4 is H or can optionally form a bond with Y3;
R5 is H, -NRbRd, or ¨0Ra;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai and Rbi are each independently H or a counterion; and
¨0W1 is OH at a pH of about 1 or OR is 0- at physiological pH.
In some embodiments, B is:
R3
N ---
- 96 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
H
0
wherein R3 is -OH, -SH, or NH2
In some embodiments, B is:
N-1\17
NN
NN
)
In some embodiments, B is:
NH2 0
e 0
+
In some embodiments, the modified nucleotides are a compound of Formula I-d:
R3
yi _______ y2 N
n\ Z _
D A
15
- 97 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
In some embodiments, the modified nucleotides, e.g., as employed in synthesis,
are a
compound selected from the group consisting of:
NH
O
0-,,,L0-0,,,LO-ovo N NH2
0 I
e oie oi
HO OH (BB- 247),
0
O 0 0 (-NH
e 0-P-O-P-O-P-0 0 NN NH2
Os 60 Oe
HO OH (BB- 248),
H3C
O 0 0
II II II
O-P-O-P-O-P-O N NH2
(Se O Oe
HO OH (BB- 249),
NH2
0 0 0 / I
eNN
Oe oe
HO OH (BB- 250),
NH2
NN
0 0 0 N3II NN
I
e
Oe oe
HO OH (BB-251),
NH
N
O 0 0 NN
O-P-O-P-O-P-O 0
6 60 Oe
HO OH (BB- 252),
- 98 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
CI
NN
O 0 0 )
11
e
Oe 6 (Se
HO OH (BB- 253),
NH
N
O 0 0
ii ii NN
e
Oa Oe Oe
HO OH (BB- 254),
0
O 0 0
II
NNNH2
Oa (Se (Se
HO OH (BB- 255),
NJNH
O 0 0 0¨K
II II
eo_p_o_p_o_p_o 0 1\1---NNH2
(Se (Se 6
HO OH (BB- 256),
0
O 0 0 NN
,1
11
O-P-O-P-O-P-O 0
Os (Se (Se
HO OH (BB- 257), and
Cl
O 0 0
1\1--N NH2
O-P-O-P-O-P-O 0
(Se (Se (Se
HO OH (BB- 258), or a pharmaceutically
acceptable salt
thereof.
- 99 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, the modified nucleotides, e.g., as employed in synthesis,
are a
compound selected from the group consisting of:
e e I
N-.......AN N-____.---=----..-N
9I
N----N 9 9 9 ____ 1 )
N----Nr
00 P 0 _____ \()/ GOPOPOPO
Oe 6 Oe Oe \cy
HO OH (BB- 259), HO OH (BB- 260),
H2N H2N
N -----\ N \
0 1 N 1 N
N 9 9 9
e II
0-p-0 0/ e0-P-O-P-0-p-0 0 N
0 e oe oe oe
HO OH (BB- 261), HO OH (BB- 262),
e e
o o
\e \e
N.,_.._N N...........N
o I 9 9 9I
N---- N N ----N
-0-F)-0-0 0-P-O-P-O-P-00
Oe Oa (Se Oe
HO OH (BB- 263), HO OH (BB- 264),
e 0
S s
\e \e
NN,....,-------õN
o
N I
(, 9 9 9 __ I
(2, , ----N N ----N
Oe -
\cyF' 0 v0i
Oe 60 6 \
i _______________________________________________________ I
HO OH (BB- 265), HO OH (BB- 266),
NH2 e NH2 0
N -,---"L N --C)
0 I
e ii N----N 9 9 9
e 1,-0-0
N
Oe Oe (Se oe N
HO OH (BB- 267), HO OH
(BB- 268),
- 100 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
H,N---,..õ,.,...-..o.-----..õ-O--õ_õ.--"-,o
N.....N
H
0 1 0()
ii
0 P 0 ____ y)f--'N
Oa
NH2
HO OH (BB- 269),
H,N(:)0()
NN
H
0 0 0 1 0
0
e0-P11-0-P-O-P-OorN
(se (se (!)e
NH2
HO OH (BB- 270),
0
0
("NH ("NH
_ 0
N--- 0 0 0
II 0 0
(0-1g-0 0 N o
Oe ¨p (se (se (se
oN<c,,) o o
(BB- 271), (BB- 272),
0 0
('NH fjcH
0
N"." 80 0 0 0
0-P-0¨ 04-0-1g-0-1g-0 N---0
Oe (se (Se
HO OH (BB- 273), HO OH (BB-
274),
0
0
A
F3CA0 HN
_-_A CF3 NH2
--
/ \ N
/ N
0 0 0 N---- 0 0 0
o 0i 0 6 1\1---0
60 6e 6e 66 6e 6e
HO OH (BB- 275), HO OH (BB- 276),
N(1H2 . NH2
/ \ N / N
0 0 0
N--- 0 0 0
,,
e
e e e
' 0-P-O-P-O-P-0¨\(0 0 60-P-O-
P11-0-Pli-00r0
6 6e 6 6 6e 6
HO OH (BB- 277), HO OH
(BB- 278),
-101 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0 0
H3C\___A
H3C- ANH NH
H3C
Z1-4N (/-4N
O 0 0
N---- e 0 0 0
N----
o-0F1,11-eoteoteo¨\(0 o eo-I1O-eo-O-eo-
O-eoo o
HO OH (BB- 279), HO OH
(BB- 280),
NH2 NH2
F2HC FH2C
Z/-4N
Z/-4N
O 0 0 II II II N---- e 011-(s-ov(s-ovo
1\1--
e 0-1-0-1-0-1-0 0 0-1:1' 1 ¨\,0 0
1 1 ¨\s,,:,0..õ(
oe oe Oe oe oe Oe
H6 OH (BB- 281), HO OH
(BB- 282),
0
F3CA
NH2 NH
H3C0
\
/ N
(/-4N
O 0 0
N---- II 0 0 0
II II II N----
e 0 o-
0-1-0-1-0-1-0 0 e -04-04-0 0
e e e
1 1 ¨\s,,;,..õ(
O o Oge (se
(se ¨\,0
H6 OH (BB- 283), HO OH
(BB- 284),
0 0
F3C0e HO
NH , 8
N----NNH
O 0 0
0 0 0 \
8 04-0 N----4-04-0 0 8 04-04-
04-0¨\(0 0
06 06 (Se Oe Oe Oe
HO OH (BB- 285), H6 OH
(BB- 286),
0
CHo
' -1(NH NH2
HN4N H3C----\N4
N
O 0 0 \ 0 0 0 \
60-P-O-P-O-P-0 0 0 6
HO OH (BB- 287), and HO OH (BB- 288),
or a pharmaceutically acceptable salt thereof.
Modifications on the Internucleoside Linkage
The modified nucleotides, which may be incorporated into a polynucleotide
molecule, can
be modified on the internucleoside linkage (e.g., phosphate backbone). Herein,
in the context of the
polynucleotide backbone, the phrases "phosphate" and "phosphodiester" are used
interchangeably.
- 102 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Backbone phosphate groups can be modified by replacing one or more of the
oxygen atoms with a
different substituent.
The modified nucleosides and nucleotides can include the wholesale replacement
of an
unmodified phosphate moiety with another internucleoside linkage as described
herein. Examples of
modified phosphate groups include, but are not limited to, phosphorothioate,
phosphoroselenates,
boranophosphates, boranophosphate esters, hydrogen phosphonates,
phosphoramidates,
phosphorodiamidates, alkyl or aryl phosphonates, and phosphotriesters.
Phosphorodithioates have
both non-linking oxygens replaced by sulfur. The phosphate linker can also be
modified by the
replacement of a linking oxygen with nitrogen (bridged phosphoramidates),
sulfur (bridged
phosphorothioates), and carbon (bridged methylene-phosphonates).
The modified nucleosides and nucleotides can include the replacement of one or
more of
the non-bridging oxygens with a borane moiety (BH3) , sulfur (thio), methyl,
ethyl and/or methoxy. As
a non-limiting example, two non-bridging oxygens at the same position (e.g.,
the alpha (a), beta (13) or
gamma (y) position) can be replaced with a sulfur (thio) and a methoxy.
The replacement of one or more of the oxygen atoms at the a position of the
phosphate
moiety (e.g., a-thio phosphate) is provided to confer stability (such as
against exonucleases and
endonucleases) to RNA and DNA through the unnatural phosphorothioate backbone
linkages.
Phosphorothioate DNA and RNA have increased nuclease resistance and
subsequently a longer half-
life in a cellular environment. While not wishing to be bound by theory,
phosphorothioate linked
polynucleotide molecules are expected to also reduce the innate immune
response through weaker
binding/activation of cellular innate immune molecules.
In specific embodiments, a modified nucleoside includes an alpha-thio-
nucleoside (e.g., 5'-
0-(1-thiophosphate)-adenosine, 5'-0-(1-thiophosphate)-cytidine (a-thio-
cytidine), 5'-0-(1-
thiophosphate)-guanosine, 5'-0-(1-thiophosphate)-uridine, or 5'-0-(1-
thiophosphate)-pseudouridine).
Other internucleoside linkages that may be employed according to the present
invention,
including internucleoside linkages which do not contain a phosphorous atom,
are described herein
below.
Combinations of Modified Sugars, Nucleobases, and Internucleoside Linkages
The polynucleotides of the invention can include a combination of
modifications to the
sugar, the nucleobase, and/or the internucleoside linkage. These combinations
can include any one
or more modifications described herein. For examples, any of the nucleotides
described herein in
Formulas (la), (la-1)-(la-3), (lb)-(10, (lia)-(lip), (lib-1), (lib-2), (lic-1)-
(lic-2), (lin-1), (lin-2), (Iva)-(Iv1),
and (Ixa)-(Ixr) can be combined with any of the nucleobases described herein
(e.g., in Formulas (b1)-
(b43) or any other described herein).
Synthesis of Polynucleotide Molecules
The polynucleotide molecules for use in accordance with the invention may be
prepared
according to any useful technique, as described herein. The modified
nucleosides and nucleotides
used in the synthesis of polynucleotide molecules disclosed herein can be
prepared from readily
available starting materials using the following general methods and
procedures. Where typical or
preferred process conditions (e.g., reaction temperatures, times, mole ratios
of reactants, solvents,
- 103 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
pressures, etc.) are provided, a skilled artisan would be able to optimize and
develop additional
process conditions. Optimum reaction conditions may vary with the particular
reactants or solvent
used, but such conditions can be determined by one skilled in the art by
routine optimization
procedures.
The processes described herein can be monitored according to any suitable
method
known in the art. For example, product formation can be monitored by
spectroscopic means, such as
nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared
spectroscopy, spectrophotometry
(e.g., UV-visible), or mass spectrometry, or by chromatography such as high
performance liquid
chromatography (HPLC) or thin layer chromatography.
Preparation of polynucleotide molecules of the present invention can involve
the protection
and deprotection of various chemical groups. The need for protection and
deprotection, and the
selection of appropriate protecting groups can be readily determined by one
skilled in the art. The
chemistry of protecting groups can be found, for example, in Greene, et al.,
Protective Groups in
Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein
by reference in its
entirety.
The reactions of the processes described herein can be carried out in suitable
solvents,
which can be readily selected by one of skill in the art of organic synthesis.
Suitable solvents can be
substantially nonreactive with the starting materials (reactants), the
intermediates, or products at the
temperatures at which the reactions are carried out, i.e., temperatures which
can range from the
solvent's freezing temperature to the solvent's boiling temperature. A given
reaction can be carried
out in one solvent or a mixture of more than one solvent. Depending on the
particular reaction step,
suitable solvents for a particular reaction step can be selected.
Resolution of racemic mixtures of modified polynucleotides or nucleic acids
(e.g.,
polynucleotides or modified mRNA molecules) can be carried out by any of
numerous methods
known in the art. An example method includes fractional recrystallization
using a "chiral resolving
acid" which is an optically active, salt-forming organic acid. Suitable
resolving agents for fractional
recrystallization methods are, for example, optically active acids, such as
the D and L forms of tartaric
acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic
acid, lactic acid or the various
optically active camphorsulfonic acids. Resolution of racemic mixtures can
also be carried out by
elution on a column packed with an optically active resolving agent (e.g.,
dinitrobenzoylphenylglycine). Suitable elution solvent composition can be
determined by one skilled
in the art.
Modified nucleosides and nucleotides (e.g., building block molecules) can be
prepared
according to the synthetic methods described in Ogata et al., J. Org. Chem.
74:2585-2588 (2009);
Purmal et al., Nucl. Acids Res. 22(1): 72-78, (1994); Fukuhara et al.,
Biochemistry, 1(4): 563-568
(1962); and Xu et al., Tetrahedron, 48(9): 1729-1740 (1992), each of which are
incorporated by
reference in their entirety.
The polynucleotides of the invention may or may not be uniformly modified
along the
entire length of the molecule. For example, one or more or all types of
nucleotide (e.g., purine or
pyrimidine, or any one or more or all of A, G, U, C) may or may not be
uniformly modified in a
polynucleotide of the invention, or in a given predetermined sequence region
thereof. In some
- 104 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
embodiments, all nucleotides X in a polynucleotide of the invention (or in a
given sequence region
thereof) are modified, wherein X may any one of nucleotides A, G, U, C, or any
one of the
combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
Different sugar modifications, nucleotide modifications, and/or
internucleoside linkages
(e.g., backbone structures) may exist at various positions in the
polynucleotide. One of ordinary skill
in the art will appreciate that the nucleotide analogs or other
modification(s) may be located at any
position(s) of a polynucleotide such that the function of the polynucleotide
is not substantially
decreased. A modification may also be a 5' or 3' terminal modification. The
polynucleotide may
contain from about 1% to about 100% modified nucleotides (either in relation
to overall nucleotide
content, or in relation to one or more types of nucleotide, i.e. any one or
more of A, G, U or C) or any
intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%,
from 1% to 60%,
from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to
20%, from 10% to
25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from
10% to 90%,
from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20%
to 60%, from
20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to
100%, from 50% to
60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from
50% to 100%,
from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80%
to 90%, from
80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95%
to 100%).
In some embodiments, the polynucleotide includes a modified pyrimidine (e.g.,
a modified
uracil/uridine/U or modified cytosine/cytidine/C). In some embodiments, the
uracil or uridine
(generally: U) in the polynucleotide molecule may be replaced with from about
1% to about 100% of a
modified uracil or modified uridine (e.g., from 1% to 20%, from 1% to 25%,
from 1% to 50%, from 1%
to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from
10% to 20%, from
10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to
80%, from 10% to
90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from
20% to 60%,
from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20%
to 100%, from
50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to
95%, from 50% to
100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%,
from 80% to 90%,
from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from
95% to 100%
of a modified uracil or modified uridine). The modified uracil or uridine can
be replaced by a
compound having a single unique structure or by a plurality of compounds
having different structures
(e.g., 2, 3, 4 or more unique structures, as described herein). In some
embodiments, the cytosine or
cytidine (generally: C) in the polynucleotide molecule may be replaced with
from about 1% to about
100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%, from
1% to 25%, from 1% to
50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1%
to 95%, from
10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to
70%, from 10% to
80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from
20% to 50%,
from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20%
to 95%, from
20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to
90%, from 50% to
95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from
70% to 100%,
from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90%
to 100%, and
- 105 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
from 95% to 100% of a modified cytosine or modified cytidine). The modified
cytosine or cytidine can
be replaced by a compound having a single unique structure or by a plurality
of compounds having
different structures (e.g., 2, 3, 4 or more unique structures, as described
herein).
In some embodiments, the present disclosure provides methods of synthesizing a
polynucleotide (e.g., the first region, first flanking region, or second
flanking region) including n
number of linked nucleosides having Formula (la-1):
_______ yi y5 u B
R3 2.
R '
Y2
Y3=P __________________
4
¨ (la-1), comprising:
a) reacting a nucleotide of Formula (IV-1):
Yl¨Y5 U
R3
N/2 y9
fl/
\ P2/m,
(IV-1),
with a phosphoramidite compound of Formula (V-1):
pi yl y5 u B
\
(R4
P3 Y2 y9
0 P2/
m'
(V-1),
wherein Y9 is H, hydroxyl, phosphoryl, pyrophosphate, sulfate, amino, thiol,
optionally
substituted amino acid, or a peptide (e.g., including from 2 to 12 amino
acids); and each P1, P2, and
P3 is, independently, a suitable protecting group; and 0 denotes a solid
support;
20
- 106 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
to provide a polynucleotide of Formula (VI-1):
Pl¨Y1¨Y5
.7E(Rzt
R
v2 y9
P2/
P3
y1 _y5
ILKIR4
R
\,2 y9
P2/m,
(VI-1), and
b) oxidizing or sulfurizing the polynucleotide of Formula (V) to yield a
polynucleotide of
Formula (VII-1):
pl yl v5
' U
./R4
R
v2 y9
F,30¨P=Y3
yl Ni5
' U
(R4
R
2 y9
P2/
(VII-1), and
c) removing the protecting groups to yield the polynucleotide of Formula (la).
In some embodiments, steps a) and b) are repeated from 1 to about 10,000
times. In
some embodiments, the methods further comprise a nucleotide selected from the
group consisting of
A, C, G and U adenosine, cytosine, guanosine, and uracil. In some embodiments,
the nucleobase
may be a pyrimidine or derivative thereof. In some embodiments, the
polynucleotide is translatable.
Other components of polynucleotides are optional, and are beneficial in some
embodiments. For example, a 5' untranslated region (UTR) and/or a 3'UTR are
provided, wherein
either or both may independently contain one or more different nucleotide
modifications. In such
embodiments, nucleotide modifications may also be present in the translatable
region. Also provided
are polynucleotides containing a Kozak sequence.
Combinations of Nucleotides
Further examples of modified nucleotides and modified nucleotide combinations
are
provided below in Table 2. These combinations of modified nucleotides can be
used to form the
- 107 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
polynucleotides of the invention. Unless otherwise noted, the modified
nucleotides may be
completely substituted for the natural nucleotides of the polynucleotides of
the invention. As a non-
limiting example, the natural nucleotide uridine may be substituted with a
modified nucleoside
described herein. In another non-limiting example, the natural nucleotide
uridine may be partially
substituted (e.g., about 0.1%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%,
50%, 55%,
60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the
modified nucleoside
disclosed herein.
Table 2: Examples of modified nucleotides and modified nucleotide
combinations.
Modified Modified Nucleotide Combination
Nucleotide
a-thio-cytidine a-thio-cytidine/5-iodo-uridine
a-thio-cytidine/N1-methyl-pseudo-uridine
a-thio-cytidine/a-thio-uridine
a-thio-cytidine/5-methyl-uridine
a-thio-cytidine/pseudo-uridine
about 50% of the cytosines are a-thio-cytidine
pseudoisocytidine pseudoisocytidine/5-iodo-uridine
pseudoisocytidine/N1-methyl-pseudouridine
pseudoisocytidine/a-thio-uridine
pseudoisocytidine/5-methyl-uridine
pseudoisocytidine/pseudouridine
about 25% of cytosines are pseudoisocytidine
pseudoisocytidine/about 50% of uridines are N1-methyl-
pseudouridine and about 50% of uridines are pseudouridine
pseudoisocytidine/about 25% of uridines are N1-methyl-
pseudouridine and about 25% of uridines are pseudouridine
(e.g., 25% N1-methyl-pseudouridine/75 /0 pseudouridine)
pyrrolo-cytidine pyrrolo-cytidine/5-iodo-uridine
pyrrolo-cytidine/N1-methyl-pseudouridine
pyrrolo-cytidine/a-thio-uridine
pyrrolo-cytidine/5-methyl-uridine
pyrrolo-cytidine/pseudouridine
about 50% of the cytosines are pyrrolo-cytidine
5-methyl-cytidine 5-methyl-cytidine/5-iodo-uridine
5-methyl-cytidine/N1-methyl-pseudouridine
5-methyl-cytidine/a-thio-uridine
5-methyl-cytidine/5-methyl-uridine
5-methyl-cytidine/pseudouridine
about 25% of cytosines are 5-methyl-cytidine
- 108 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Modified Modified Nucleotide Combination
Nucleotide
about 50% of cytosines are 5-methyl-cytidine
5-methyl-cytidine/5-methoxy-uridine
5-methyl-cytidine/5-bromo-uridine
5-methyl-cytidine/2-thio-uridine
5-methyl-cytidine/about 50% of uridines are 2-thio-uridine
about 50% of uridines are 5-methyl-cytidine/ about 50% of uridines
are 2-thio-uridine
N4-acetyl-cytidine N4-acetyl-cytidine/5-iodo-uridine
N4-acetyl-cytidine /N1-methyl-pseudouridine
N4-acetyl-cytidine /a-thio-uridine
N4-acetyl-cytidine /5-methyl-uridine
N4-acetyl-cytidine /pseudouridine
about 50% of cytosines are N4-acetyl-cytidine
about 25% of cytosines are N4-acetyl-cytidine
N4-acetyl-cytidine /5-methoxy-uridine
N4-acetyl-cytidine /5-bromo-uridine
N4-acetyl-cytidine/2-thio-uridine
about 50% of cytosines are N4-acetyl-cytidine/ about 50% of uridines
are 2-thio-uridine
5-methoxy-uridine 5-methoxy-uridine/cytidine
5-methoxy-uridine/5-methyl-cytidine
5-methoxy-uridine/5-trifluoromethyl-cytidine
5-methoxy-uridine/5-hydroxymethyl-cytidine
5-methoxy-uridine/5-bromo-cytidine
5-methoxy-uridine/ a-thio-cytidine
5-methoxy-uridine/N4-acetyl-cytidine
5-methoxy-uridine/pseudoisocytidine
about 100% of uridines are 5-methoxy-uridine
about 75% of uridines are 5-methoxy-uridine
about 50% of uridines are 5-methoxy-uridine
about 25% of uridines are 5-methoxy-uridine
Certain modified nucleotides and nucleotide combinations have been explored.
These
findings are described in U.S. Provisional Application No 61/404,413, U.S.
Patent Application No
13/251,840, U.S. Patent Application No 13/481,127, International Patent
Publication No
W02012045075, U.S. Patent Publication No U520120237975, and International
Patent Publication
No W02012045082.
- 109 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Further examples of modified nucleotide combinations are provided below in
Table 3.
These combinations of modified nucleotides can be used to form the
polynucleotides of the invention.
Table 3: Examples of modified nucleotide combinations.
Modified Nucleotide Modified Nucleotide Combination
modified cytidine having one or modified cytidine with (b10)/pseudouridine
more nucleobases of Formula (b10) modified cytidine with (b10)/N1-methyl-
pseudouridine
modified cytidine with (b10)/5-methoxy-uridine
modified cytidine with (b10)/5-methyl-uridine
modified cytidine with (b10)/5-bromo-uridine
modified cytidine with (b10)/2-thio-uridine
about 50% of cytidine substituted with modified cytidine
(b10)/ about 50% of uridines are 2-thio-uridine
modified cytidine having one or modified cytidine with (b32)/pseudouridine
more nucleobases of Formula (b32) modified cytidine with (b32)/N1-methyl-
pseudouridine
modified cytidine with (b32)/5-methoxy-uridine
modified cytidine with (b32)/5-methyl-uridine
modified cytidine with (b32)/5-bromo-uridine
modified cytidine with (b32)/2-thio-uridine
about 50% of cytidine substituted with modified cytidine
(b32)/ about 50% of uridines are 2-thio-uridine
modified uridine having one or more modified uridine with (b1)/ N4-acetyl-
cytidine
nucleobases of Formula (b1) modified uridine with (b1)/ 5-methyl-
cytidine
modified uridine having one or more modified uridine with (b8)/ N4-acetyl-
cytidine
nucleobases of Formula (b8) modified uridine with (b8)/ 5-methyl-
cytidine
modified uridine having one or more modified uridine with (b28)/ N4-acetyl-
cytidine
nucleobases of Formula (b28) modified uridine with (b28)/ 5-methyl-
cytidine
modified uridine having one or more modified uridine with (b29)/ N4-acetyl-
cytidine
nucleobases of Formula (b29) modified uridine with (b29)/ 5-methyl-
cytidine
modified uridine having one or more modified uridine with (b30)/ N4-acetyl-
cytidine
nucleobases of Formula (b30) modified uridine with (b30)/ 5-methyl-
cytidine
In some embodiments, at least 25% of the cytosines are replaced by a compound
of
Formula (b10)-(b14), (b24), (b25), or (b32)-(b35) (e.g., at least about 30%,
at least about 35%, at
least about 40%, at least about 45%, at least about 50%, at least about 55%,
at least about 60%, at
least about 65%, at least about 70%, at least about 75%, at least about 80%,
at least about 85%, at
least about 90%, at least about 95%, or about 100% of, e.g., a compound of
Formula (b10) or (b32)).
In some embodiments, at least 25% of the uracils are replaced by a compound of
Formula
(b1)-(b9), (b21)-(b23), or (b28)-(b31) (e.g., at least about 30%, at least
about 35%, at least about
40%, at least about 45%, at least about 50%, at least about 55%, at least
about 60%, at least about
65%, at least about 70%, at least about 75%, at least about 80%, at least
about 85%, at least about
- 110 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
90%, at least about 95%, or about 100% of, e.g., a compound of Formula (b1),
(b8), (b28), (b29), or
(b30)).
In some embodiments, at least 25% of the cytosines are replaced by a compound
of
Formula (b10)-(b14), (b24), (b25), or (b32)-(b35) (e.g. Formula (b10) or
(b32)), and at least 25% of
the uracils are replaced by a compound of Formula (b1)-(b9), (b21)-(b23), or
(b28)-(b31) (e.g.
Formula (b1), (b8), (b28), (b29), or (b30)) (e.g., at least about 30%, at
least about 35%, at least about
40%, at least about 45%, at least about 50%, at least about 55%, at least
about 60%, at least about
65%, at least about 70%, at least about 75%, at least about 80%, at least
about 85%, at least about
90%, at least about 95%, or about 100%).
Modifications including Linker and a Payload
The nucleobase of the nucleotide can be covalently linked at any chemically
appropriate
position to a payload, e.g., detectable agent or therapeutic agent. For
example, the nucleobase can
be deaza-adenosine or deaza-guanosine and the linker can be attached at the C-
7 or C-8 positions of
the deaza-adenosine or deaza-guanosine. In other embodiments, the nucleobase
can be cytosine or
uracil and the linker can be attached to the N-3 or C-5 positions of cytosine
or uracil. Scheme 1
below depicts an exemplary modified nucleotide wherein the nucleobase,
adenine, is attached to a
linker at the C-7 carbon of 7-deaza adenine. In addition, Scheme 1 depicts the
modified nucleotide
with the linker and payload, e.g., a detectable agent, incorporated onto the
3' end of the mRNA.
Disulfide cleavage and 1,2-addition of the thiol group onto the propargyl
ester releases the detectable
agent. The remaining structure (depicted, for example, as pApC5Parg in Scheme
1) is the inhibitor.
The rationale for the structure of the modified nucleotides is that the
tethered inhibitor sterically
interferes with the ability of the polymerase to incorporate a second base.
Thus, it is critical that the
tether be long enough to affect this function and that the inhibiter be in a
stereochemical orientation
that inhibits or prohibits second and follow on nucleotides into the growing
polynucleotide strand.
-111-

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Scheme 1
o3s 0 i id so3H
--- --- ¨ 1
-
Ne N 111,
C
HN 0
NH2 H ,,...---,..õõ---.A
Y's NH2
NI ' \ 0 0
I NH2
N N
A Capless pCpC5 Parg ' N
9 9 9 0 I
N 0
-OPOPOPO
K -0. P 0
OH OH 1:1-,0,------0
0j=õ0
1
incorporation Cy5
0
NH HN 0
N H
------/=v0
S¨Sr
RNA ¨11 N H3
N N 0
0
0 _______________________________________________ i NH
Oj
/
N
I
OH OH N 0
Cleavage of S-S bond , 0
(:)
P, c7f
- _c5 o
/
NH2
N , \ = (DISH P'
-0- \
RNA-1¨ I 0-
1\1----N 0
0
________________ 0)
\
OH OH
V
NH2
N
RNA-1¨ I ' 0
N N
0 S
+ \-
cC)
OH OH
- 112 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Linker
The term "linker" as used herein refers to a group of atoms, e.g., 10-1,000
atoms, and can
be comprised of the atoms or groups such as, but not limited to, carbon,
amino, alkylamino, oxygen,
sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached
to a modified nucleoside or
nucleotide on the nucleobase or sugar moiety at a first end, and to a payload,
e.g., detectable or
therapeutic agent, at a second end. The linker is of sufficient length as to
not interfere with
incorporation into a nucleic acid sequence.
Examples of chemical groups that can be incorporated into the linker include,
but are not
limited to, an alkyl, alkene, an alkyne, an amido, an ether, a thioether, an
or an ester group. The
linker chain can also comprise part of a saturated, unsaturated or aromatic
ring, including polycyclic
and heteroaromatic rings wherein the heteroaromatic ring is an aryl group
containing from one to four
heteroatoms, N, 0 or S. Specific examples of linkers include, but are not
limited to, unsaturated
alkanes, polyethylene glycols, and dextran polymers.
For example, the linker can include ethylene or propylene glycol monomeric
units, e.g.,
diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene
glycol, tetraethylene glycol, or
tetraethylene glycol. In some embodiments, the linker can include a divalent
alkyl, alkenyl, and/or
alkynyl moiety. The linker can include an ester, amide, or ether moiety.
Other examples include cleavable moieties within the linker, such as, for
example, a
disulfide bond (-S-S-) or an azo bond (-N=N-), which can be cleaved using a
reducing agent or
photolysis. A cleavable bond incorporated into the linker and attached to a
modified nucleotide, when
cleaved, results in, for example, a short "scar" or chemical modification on
the nucleotide. For
example, after cleaving, the resulting scar on a nucleotide base, which formed
part of the modified
nucleotide, and is incorporated into a polynucleotide strand, is unreactive
and does not need to be
chemically neutralized. This increases the ease with which a subsequent
nucleotide can be
incorporated during sequencing of a nucleic acid polymer template. For
example, conditions include
the use of tris(2-carboxyethyl)phosphine (TCEP), dithiothreitol (DTT) and/or
other reducing agents for
cleavage of a disulfide bond. A selectively severable bond that includes an
amido bond can be
cleaved for example by the use of TCEP or other reducing agents, and/or
photolysis. A selectively
severable bond that includes an ester bond can be cleaved for example by
acidic or basic hydrolysis.
Payload
The methods and compositions described herein are useful for delivering a
payload to a
biological target. The payload can be used, e.g., for labeling (e.g., a
detectable agent such as a
fluorophore), or for therapeutic purposes (e.g., a cytotoxin or other
therapeutic agent).
Payload: Therapeutic Agents
In some embodiments the payload is a therapeutic agent such as a cytotoxin,
radioactive
ion, chemotherapeutic, or other therapeutic agent. A cytotoxin or cytotoxic
agent includes any agent
that is detrimental to cells. Examples include taxol, cytochalasin B,
gramicidin D, ethidium bromide,
emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine,
colchicin, doxorubicin,
daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin,
actinomycin D, 1-
dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine,
propranolol, puromycin,
- 113 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
maytansinoids, e.g., maytansinol (see U.S. Pat. No. 5,208,020), CC-1065 (see
U.S. Pat. Nos.
5,475,092, 5,585,499, 5,846,545) and analogs or homologs thereof. Radioactive
ions include, but are
not limited to iodine (e.g., iodine 125 or iodine 131), strontium 89,
phosphorous, palladium, cesium,
iridium, phosphate, cobalt, yttrium 90, Samarium 153 and praseodymium. Other
therapeutic agents
include, but are not limited to, antimetabolites (e.g., methotrexate, 6-
mercaptopurine, 6-thioguanine,
cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g.,
mechlorethamine, thioepa
chlorambucil, CC-1065, melphalan, carmustine (BSNU) and lomustine (CCNU),
cyclothosphamide,
busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-
dichlorodiamine platinum (II) (DDP)
cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and
doxorubicin), antibiotics (e.g.,
dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin
(AMC)), and anti-
mitotic agents (e.g., vincristine, vinblastine, taxol and maytansinoids).
Payload: Detectable Agents
Examples of detectable substances include various organic small molecules,
inorganic
compounds, nanoparticles, enzymes or enzyme substrates, fluorescent materials,
luminescent
materials, bioluminescent materials, chemiluminescent materials, radioactive
materials, and contrast
agents. Such optically-detectable labels include for example, without
limitation, 4-acetamido-4'-
isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives:
acridine, acridine isothiocyanate;
5-(2'-aminoethyDaminonaphthalene-1 -sulfonic acid (EDANS); 4-amino-N-[3-
vinylsulfonyl)phenylmaphthalimide-3,5 disulfonate; N-(4-anilino-l-
naphthyl)maleimide; anthranilamide;
BODIPY; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4-
methylcoumarin (AMC,
Coumarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran 151); cyanine
dyes; cyanosine; 4',6-
diaminidino-2-phenylindole (DAPI); 5' 5"-dibromopyrogallol-sulfonaphthalein
(Bromopyrogallol Red);
7-diethylamino-3-(4'-isothiocyanatophenyI)-4-methylcoumarin;
diethylenetriamine pentaacetate; 4,4'-
diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4'-
diisothiocyanatostilbene-2,2'-disulfonic acid;
5-[dimethylamino]-naphthalene-1-sulfonyl chloride (DNS, dansylchloride); 4-
dimethylaminophenylazopheny1-4'-isothiocyanate (DABITC); eosin and
derivatives; eosin, eosin
isothiocyanate, erythrosin and derivatives; erythrosin B, erythrosin,
isothiocyanate; ethidium;
fluorescein and derivatives; 5-carboxyfluorescein (FAM), 5-(4,6-
dichlorotriazin-2-yDaminofluorescein
(DTAF), 2',7'-dimethoxy-4'5'-dichloro-6-carboxyfluorescein, fluorescein,
fluorescein isothiocyanate,
QFITC, (XRITC); fluorescamine; IR144; IR1446; Malachite Green isothiocyanate;
4-
methylumbelliferoneortho cresolphthalein; nitrotyrosine; pararosaniline;
Phenol Red; B-phycoerythrin;
o-phthaldialdehyde; pyrene and derivatives: pyrene, pyrene butyrate,
succinimidyl 1-pyrene;
butyrate quantum dots; Reactive Red 4 (CibacronTM Brilliant Red 3B-A)
rhodamine and derivatives:
6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B
sulfonyl chloride
rhodarnine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate,
sulforhodamine B,
sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas
Red);
N,N,N',Ntetramethy1-6-carboxyrhodamine (TAMRA); tetramethyl rhodamine;
tetramethyl rhodamine
isothiocyanate (TRITC); riboflavin; rosolic acid; terbium chelate derivatives;
Cyanine-3 (Cy3);
Cyanine-5 (Cy5); Cyanine-5.5 (Cy5.5), Cyanine-7 (Cy7); IRD 700; IRD 800; Alexa
647; La Jolta Blue;
- 114 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
phthalo cyanine; and naphthalo cyanine. In some embodiments, the detectable
label is a fluorescent
dye, such as Cy5 and Cy3.
Examples luminescent material includes luminol; examples of bioluminescent
materials
include luciferase, luciferin, and aequorin.
Examples of suitable radioactive material include 18F567Ga, aimKr, 82Rb,
12315 133xe,
201T1, 1251, 35S, 14C,
3 99m
T1, 1, S, C, or H, Tc (e.g., as pertechnetate (technetate(VII), Tc04)
either directly or
indirectly, or other radioisotope detectable by direct counting of
radioemission or by scintillation
counting.
In addition, contrast agents, e.g., contrast agents for MRI or NMR, for X-ray
CT, Raman
imaging, optical coherence tomography, absorption imaging, ultrasound imaging,
or thermal imaging
can be used. Exemplary contrast agents include gold (e.g., gold
nanoparticles), gadolinium (e.g.,
chelated Gd), iron oxides (e.g., superparamagnetic iron oxide (SP10),
monocrystalline iron oxide
nanoparticles (MIONs), and ultrasmall superparamagnetic iron oxide (USP10)),
manganese chelates
(e.g., Mn-DPDP), barium sulfate, iodinated contrast media (iohexol),
microbubbles, or
perfluorocarbons can also be used.
In some embodiments, the detectable agent is a non-detectable pre-cursor that
becomes
detectable upon activation. Examples include fluorogenic tetrazine-fluorophore
constructs (e.g.,
tetrazine-BODIPY FL, tetrazine-Oregon Green 488, or tetrazine-BODIPY TMR-X) or
enzyme
activatable fluorogenic agents (e.g., PROSENSE (VisEn Medical)).
When the compounds are enzymatically labeled with, for example, horseradish
peroxidase, alkaline phosphatase, or luciferase, the enzymatic label is
detected by determination of
conversion of an appropriate substrate to product.
In vitro assays in which these compositions can be used include enzyme linked
immunosorbent assays (ELISAs), immunoprecipitations, immunofluorescence,
enzyme immunoassay
(EIA), radioimmunoassay (RIA), and Western blot analysis.
Labels other than those described herein are contemplated by the present
disclosure,
including other optically-detectable labels. Labels can be attached to the
modified nucleotide of the
present disclosure at any position using standard chemistries such that the
label can be removed
from the incorporated base upon cleavage of the cleavable linker.
Payload:Cell Penetrating Payloads
In some embodiments, the modified nucleotides and modified nucleic acids can
also
include a payload that can be a cell penetrating moiety or agent that enhances
intracellular delivery of
the compositions. For example, the compositions can include a cell-penetrating
peptide sequence
that facilitates delivery to the intracellular space, e.g., HIV-derived TAT
peptide, penetratins,
transportans, or hCT derived cell-penetrating peptides, see, e.g., Caron et
al., (2001) Mol Ther.
3(3):310-8; Lange!, Cell-Penetrating Peptides: Processes and Applications (CRC
Press, Boca Raton
FL 2002); El-Andaloussi et al., (2005) Curr Pharm Des. 11(28):3597-611; and
Deshayes et al., (2005)
Cell Mol Life Sci. 62(16):1839-49. The compositions can also be formulated to
include a cell
penetrating agent, e.g., liposomes, which enhance delivery of the compositions
to the intracellular
space.
- 115 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Payload:Biological Targets
The modified nucleotides and modified nucleic acids described herein can be
used to
deliver a payload to any biological target for which a specific ligand exists
or can be generated. The
ligand can bind to the biological target either covalently or non-covalently.
Exemplary biological targets include biopolymers, e.g., antibodies, nucleic
acids such as
RNA and DNA, proteins, enzymes; exemplary proteins include enzymes, receptors,
and ion channels.
In some embodiments the target is a tissue- or cell-type specific marker,
e.g., a protein that is
expressed specifically on a selected tissue or cell type. In some embodiments,
the target is a
receptor, such as, but not limited to, plasma membrane receptors and nuclear
receptors; more
specific examples include G-protein-coupled receptors, cell pore proteins,
transporter proteins,
surface-expressed antibodies, HLA proteins, MHC proteins and growth factor
receptors.
Synthesis of Modified Nucleotides
The modified nucleosides and nucleotides disclosed herein can be prepared from
readily
available starting materials using the following general methods and
procedures. It is understood that
where typical or preferred process conditions (i.e., reaction temperatures,
times, mole ratios of
reactants, solvents, pressures, etc.) are given; other process conditions can
also be used unless
otherwise stated. Optimum reaction conditions may vary with the particular
reactants or solvent used,
but such conditions can be determined by one skilled in the art by routine
optimization procedures.
The processes described herein can be monitored according to any suitable
method
known in the art. For example, product formation can be monitored by
spectroscopic means, such as
nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared
spectroscopy, spectrophotometry
(e.g., UV-visible), or mass spectrometry, or by chromatography such as high
performance liquid
chromatography (HPLC) or thin layer chromatography.
Preparation of modified nucleosides and nucleotides can involve the protection
and
deprotection of various chemical groups. The need for protection and
deprotection, and the selection
of appropriate protecting groups can be readily determined by one skilled in
the art. The chemistry of
protecting groups can be found, for example, in Greene, et al., Protective
Groups in Organic
Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by
reference in its entirety.
The reactions of the processes described herein can be carried out in suitable
solvents,
which can be readily selected by one of skill in the art of organic synthesis.
Suitable solvents can be
substantially nonreactive with the starting materials (reactants), the
intermediates, or products at the
temperatures at which the reactions are carried out, i.e., temperatures which
can range from the
solvent's freezing temperature to the solvent's boiling temperature. A given
reaction can be carried
out in one solvent or a mixture of more than one solvent. Depending on the
particular reaction step,
suitable solvents for a particular reaction step can be selected.
Resolution of racemic mixtures of modified nucleosides and nucleotides can be
carried out
by any of numerous methods known in the art. An example method includes
fractional
recrystallization using a "chiral resolving acid" which is an optically
active, salt-forming organic acid.
Suitable resolving agents for fractional recrystallization methods are, for
example, optically active
- 116 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
acids, such as the D and L forms of tartaric acid, diacetyltartaric acid,
dibenzoyltartaric acid, mandelic
acid, malic acid, lactic acid or the various optically active camphorsulfonic
acids. Resolution of
racemic mixtures can also be carried out by elution on a column packed with an
optically active
resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent
composition can be
determined by one skilled in the art.
Modified Nucleic Acids
The present disclosure provides nucleic acids (or polynucleotides), including
RNAs such
as mRNAs that contain one or more modified nucleosides (termed "modified
nucleic acids") or
nucleotides as described herein, which have useful properties including the
lack of a substantial
induction of the innate immune response of a cell into which the mRNA is
introduced. Because these
modified nucleic acids enhance the efficiency of protein production,
intracellular retention of nucleic
acids, and viability of contacted cells, as well as possess reduced
immunogenicity, these nucleic
acids having these properties are also termed "enhanced nucleic acids" herein.
The term "nucleic acid," in its broadest sense, includes any compound and/or
substance
that is or can be incorporated into an oligonucleotide chain. In this context,
the term nucleic acid is
used synonymously with polynucleotide. Exemplary nucleic acids for use in
accordance with the
present disclosure include, but are not limited to, one or more of DNA, RNA
including messenger
mRNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs,
shRNAs, miRNAs,
antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix
formation, aptamers,
vectors, etc., described in detail herein.
Provided are modified nucleic acids containing a translatable region and one,
two, or more
than two different nucleoside modifications. In some embodiments, the modified
nucleic acid exhibits
reduced degradation in a cell into which the nucleic acid is introduced,
relative to a corresponding
unmodified nucleic acid. Exemplary nucleic acids include ribonucleic acids
(RNAs), deoxyribonucleic
acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), or a
hybrid thereof. In
preferred embodiments, the modified nucleic acid includes messenger RNAs
(mRNAs). As described
herein, the nucleic acids of the present disclosure do not substantially
induce an innate immune
response of a cell into which the mRNA is introduced.
In certain embodiments, it is desirable to intracellularly degrade a modified
nucleic acid
introduced into the cell, for example if precise timing of protein production
is desired. Thus, the
present disclosure provides a modified nucleic acid containing a degradation
domain, which is
capable of being acted on in a directed manner within a cell.
Other components of nucleic acid are optional, and are beneficial in some
embodiments.
For example, a 5' untranslated region (UTR) and/or a 3'UTR are provided,
wherein either or both may
independently contain one or more different nucleoside modifications. In such
embodiments,
nucleoside modifications may also be present in the translatable region. Also
provided are nucleic
acids containing a Kozak sequence.
Additionally, provided are nucleic acids containing one or more intronic
nucleotide
sequences capable of being excised from the nucleic acid.
-117-

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Further, provided are nucleic acids containing an internal ribosome entry site
(IRES). An
IRES may act as the sole ribosome binding site, or may serve as one of
multiple ribosome binding
sites of an mRNA. An mRNA containing more than one functional ribosome binding
site may encode
several peptides or polypeptides that are translated independently by the
ribosomes ("multicistronic
mRNA"). When nucleic acids are provided with an IRES, further optionally
provided is a second
translatable region. Examples of IRES sequences that can be used according to
the present
disclosure include without limitation, those from picornaviruses (e.g. FMDV),
pest viruses (CFFV),
polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and-mouth
disease viruses (FMDV),
hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine
leukemia virus (MLV), simian
immune deficiency viruses (Sly) or cricket paralysis viruses (CrPV).
In some embodiments, the nucleic acid is a compound of Formula Xl-a:
X
_p_ B
6Rcl
y2 A
1-141¨P=X
ORcl
Xl-a
wherein:
denotes an optional double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or -CRaRb- when denotes a single bond, or U is -CRa-
when
denotes a double bond;
A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, or a
peptide
comprising 2 to 12 amino acids;
X is 0 or S;
bl, l
i, _NRaR
each of Y1 is independently selected from _oRa and ¨SRai ;
each of Y2 and Y3 are independently selected from 0, -CRaRb-, NRc, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai is H;
_OR cl is OH at a pH of about 1 or ¨ORcl is 0- at physiological pH; and
B is nucleobase;
provided that the ring encompassing the variables A, B, U, Y2 and Y3 cannot be
ribose.
- 118 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, B is a nucleobase of Formula XII-a, XII-b, or XII-c:
R2 0 R2
R1V 'N
, R4 R3,N)-NH R3,N
'-
-w-Lx 0
XII-a XII-b XII-c
wherein:
denotes a single or double bond;
X is 0 or S;
U and W are each independently CH or N;
V is 0, S, C or N;
wherein when V is C then R1 is H, C1_6 alkyl, C2_6 alkenyl, C2_6 alkynyl,
halo, or ¨OW,
wherein C1_6 alkyl, C2_6 alkenyl, and C2_6 alkynyl are each optionally
substituted with ¨OH, -NRaRb, -
SH, -C(0)R, -C(0)OR, -NHC(0)Rc, or -NHC(0)0Rc;
and wherein when V is 0, S, or N then R1 is absent;
R2 is H, -SRc, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are
attached
can form a 5- or 6-membered ring optionally substituted with 1-4 substituents
selected from halo, -
OH, -SH, -NRaRb, C1_20 alkyl, C2_20 alkenyl, C2_20 alkynyl, C1_20 alkoxy, or
C1_20 thioalkyl;
R3 is H or C1_20 alkyl;
R4 is H or C1_20 alkyl; wherein when denotes a double bond then R4 is
absent, or N-R4,
taken together, forms a positively charged N substituted with C1_20 alkyl;
Ra and Rb are each independently H, C1_20 alkyl, C2_20 alkenyl, C2_20 alkynyl,
or C6_20 aryl;
and
Rc is H, C1_20 alkyl, C2_20 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula XII-al XII-a2, XII-a3, XII-
a4, or XII-
a5:
X R2 R2 R2 0
R1N,R4
NN.R4 RN R1 RyL
N NH
'0 X N 0 'N
XII-al XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In some embodiments, the nucleic acid contains a plurality of structurally
unique
compounds of Formula Xl-a.
In some embodiments, at least 25% of the cytosines are replaced by a compound
of
Formula Xl-a (e.g., at least about 30%, at least about 35%, at least about
40%, at least about 45%, at
least about 50%, at least about 55%, at least about 60%, at least about 65%,
at least about 70%, at
- 119 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
least about 75%, at least about 80%, at least about 85%, at least about 90%,
at least about 95%, or
about 100%).
In some embodiments, at least 25% of the uracils are replaced by a compound of
Formula
Xl-a (e.g., at least about 30%, at least about 35%, at least about 40%, at
least about 45%, at least
about 50%, at least about 55%, at least about 60%, at least about 65%, at
least about 70%, at least
about 75%, at least about 80%, at least about 85%, at least about 90%, at
least about 95%, or about
100%).
In some embodiments, at least 25% of the cytosines and 25% of the uracils are
replaced
by a compound of Formula Xl-a (e.g., at least about 30%, at least about 35%,
at least about 40%, at
least about 45%, at least about 50%, at least about 55%, at least about 60%,
at least about 65%, at
least about 70%, at least about 75%, at least about 80%, at least about 85%,
at least about 90%, at
least about 95%, or about 100%).
In some embodiments, the nucleic acid is translatable.
In some embodiments, when the nucleic acid includes a nucleotide modified with
a linker
and payload, for example, as described herein, the nucleotide modified with a
linker and payload is
on the 3' end of the nucleic acid.
Major Groove Interacting Partners
As described herein, the phrase "major groove interacting partner" refers RNA
recognition
receptors that detect and respond to RNA ligands through interactions, e.g.
binding, with the major
groove face of a nucleotide or nucleic acid. As such, RNA ligands comprising
modified nucleotides or
nucleic acids as described herein decrease interactions with major groove
binding partners, and
therefore decrease an innate immune response, or expression and secretion of
pro-inflammatory
cytokines, or both.
Example major groove interacting, e.g. binding, partners include, but are not
limited to the
following nucleases and helicases. Within membranes, TLRs (Toll-like
Receptors) 3, 7, and 8 can
respond to single- and double-stranded RNAs. Within the cytoplasm, members of
the superfamily 2
class of DEX(D/H) helicases and ATPases can sense RNAs to initiate antiviral
responses. These
helicases include the RIG-I (retinoic acid-inducible gene I) and MDA5
(melanoma differentiation-
associated gene 5). Other examples include laboratory of genetics and
physiology 2 (LGP2), HIN-
200 domain containing proteins, or Helicase-domain containing proteins.
- 120 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Prevention or reduction of innate cellular immune response
The term "innate immune response" includes a cellular response to exogenous
single
stranded nucleic acids, generally of viral or bacterial origin, which involves
the induction of cytokine
expression and release, particularly the interferons, and cell death. Protein
synthesis is also reduced
during the innate cellular immune response. While it is advantageous to
eliminate the innate immune
response in a cell which is triggered by introduction of exogenous nucleic
acids, the present
disclosure provides modified nucleic acids such as mRNAs that substantially
reduce the immune
response, including interferon signaling, without entirely eliminating such a
response. In some
embodiments, the immune response is reduced by 10%, 20%, 30%, 40%, 50%, 60%,
70%, 80%,
90%, 95%, 99%, 99.9%, or greater than 99.9% as compared to the immune response
induced by a
corresponding unmodified nucleic acid. Such a reduction can be measured by
expression or activity
level of Type 1 interferons or the expression of interferon-regulated genes
such as the toll-like
receptors (e.g., TLR7 and TLR8). Reduction or lack of induction of innate
immune response can also
be measured by decreased cell death following one or more administrations of
modified RNAs to a
cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or
over 95% less than the
cell death frequency observed with a corresponding unmodified nucleic acid.
Moreover, cell death
may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer
than 0.01% of cells
contacted with the modified nucleic acids.
In some embodiments, the modified nucleic acids, including polynucleotides
and/or mRNA
molecules are modified in such a way as to not induce, or induce only
minimally, an immune
response by the recipient cell or organism. Such evasion or avoidance of an
immune response trigger
or activation is a novel feature of the modified polynucleotides of the
present invention.
The present disclosure provides for the repeated introduction (e.g.,
transfection) of
modified nucleic acids into a target cell population, e.g., in vitro, ex vivo,
or in vivo. The step of
contacting the cell population may be repeated one or more times (such as two,
three, four, five or
more than five times). In some embodiments, the step of contacting the cell
population with the
modified nucleic acids is repeated a number of times sufficient such that a
predetermined efficiency of
protein translation in the cell population is achieved. Given the reduced
cytotoxicity of the target cell
population provided by the nucleic acid modifications, such repeated
transfections are achievable in a
diverse array of cell types in vitro and/or in vivo.
Polypeptide variants
Provided are nucleic acids that encode variant polypeptides, which have a
certain identity
with a reference polypeptide sequence. The term "identity" as known in the
art, refers to a relationship
between the sequences of two or more peptides, as determined by comparing the
sequences. In the
art, "identity" also means the degree of sequence relatedness between
peptides, as determined by
the number of matches between strings of two or more amino acid residues.
"Identity" measures the
percent of identical matches between the smaller of two or more sequences with
gap alignments (if
any) addressed by a particular mathematical model or computer program (i.e.,
"algorithms"). Identity
of related peptides can be readily calculated by known methods. Such methods
include, but are not
limited to, those described in Computational Molecular Biology, Lesk, A. M.,
ed., Oxford University
- 121 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith,
D. W., ed.,
Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1,
Griffin, A. M., and
Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in
Molecular Biology, von
Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and
Devereux, J., eds.,
M. Stockton Press, New York, 1991; and Carillo et al., SIAM J. Applied Math.
48, 1073 (1988).
In some embodiments, the polypeptide variant has the same or a similar
activity as the
reference polypeptide. Alternatively, the variant has an altered activity
(e.g., increased or decreased)
relative to a reference polypeptide. Generally, variants of a particular
polynucleotide or polypeptide of
the present disclosure will have at least about 40%, 45%, 50%, 55%, 60%, 65%,
70%, 75%, 80%,
85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence
identity to that
particular reference polynucleotide or polypeptide as determined by sequence
alignment programs
and parameters described herein and known to those skilled in the art.
As recognized by those skilled in the art, protein fragments, functional
protein domains,
and homologous proteins are also considered to be within the scope of this
present disclosure. For
example, provided herein is any protein fragment of a reference protein
(meaning a polypeptide
sequence at least one amino acid residue shorter than a reference polypeptide
sequence but
otherwise identical) 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than
100 amino acids in length
In another example, any protein that includes a stretch of about 20, about 30,
about 40, about 50, or
about 100 amino acids which are about 40%, about 50%, about 60%, about 70%,
about 80%, about
90%, about 95%, or about 100% identical to any of the sequences described
herein can be utilized in
accordance with the present disclosure. In certain embodiments, a protein
sequence to be utilized in
accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or
more mutations as shown
in any of the sequences provided or referenced herein.
Polypeptide libraries
Also provided are polynucleotide libraries containing nucleoside
modifications, wherein the
polynucleotides individually contain a first nucleic acid sequence encoding a
polypeptide, such as an
antibody, protein binding partner, scaffold protein, and other polypeptides
known in the art.
Preferably, the polynucleotides are mRNA in a form suitable for direct
introduction into a target cell
host, which in turn synthesizes the encoded polypeptide.
In certain embodiments, multiple variants of a protein, each with different
amino acid
modification(s), are produced and tested to determine the best variant in
terms of pharmacokinetics,
stability, biocompatibility, and/or biological activity, or a biophysical
property such as expression level.
6
Such a library may contain 10, 102, iO3, iO4, i05, 10, i07, 108, i09, or over
109 possible variants
(including substitutions, deletions of one or more residues, and insertion of
one or more residues).
Polypeptide-nucleic acid complexes
Proper protein translation involves the physical aggregation of a number of
polypeptides
and nucleic acids associated with the mRNA. Provided by the present disclosure
are protein-nucleic
acid complexes, containing a translatable mRNA having one or more nucleoside
modifications (e.g.,
at least two different nucleoside modifications) and one or more polypeptides
bound to the mRNA.
- 122 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Generally, the proteins are provided in an amount effective to prevent or
reduce an innate immune
response of a cell into which the complex is introduced.
Untranslatable modified nucleic acids
As described herein, provided are mRNAs having sequences that are
substantially not
translatable. Such mRNA is effective as a vaccine when administered to a
mammalian subject.
Also provided are modified nucleic acids that contain one or more noncoding
regions.
Such modified nucleic acids are generally not translated, but are capable of
binding to and
sequestering one or more translational machinery component such as a ribosomal
protein or a
transfer RNA (tRNA), thereby effectively reducing protein expression in the
cell. The modified nucleic
acid may contain a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small
interfering RNA
(siRNA) or Piwi-interacting RNA (piRNA).
Synthesis of Modified Nucleic Acids
Nucleic acids for use in accordance with the present disclosure may be
prepared
according to any available technique including, but not limited to chemical
synthesis, enzymatic
synthesis, which is generally termed in vitro transcription, enzymatic or
chemical cleavage of a longer
precursor, etc. Methods of synthesizing RNAs are known in the art (see, e.g.,
Gait, M.J. (ed.)
Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire],
Washington, DC: IRL Press,
1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and
applications, Methods in
Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005;
both of which are
incorporated herein by reference).
Modified nucleic acids need not be uniformly modified along the entire length
of the
molecule. Different nucleotide modifications and/or backbone structures may
exist at various
positions in the nucleic acid. One of ordinary skill in the art will
appreciate that the nucleotide analogs
or other modification(s) may be located at any position(s) of a nucleic acid
such that the function of
the nucleic acid is not substantially decreased. A modification may also be a
5' or 3' terminal
modification. The nucleic acids may contain at a minimum one and at maximum
100% modified
nucleotides, or any intervening percentage, such as at least 5% modified
nucleotides, at least 10%
modified nucleotides, at least 25% modified nucleotides, at least 50% modified
nucleotides, at least
80% modified nucleotides, or at least 90% modified nucleotides. For example,
the nucleic acids may
contain a modified pyrimidine such as uracil or cytosine. In some embodiments,
at least 5%, at least
10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the
uracil in the nucleic acid is
replaced with a modified uracil. The modified uracil can be replaced by a
compound having a single
unique structure, or can be replaced by a plurality of compounds having
different structures (e.g., 2,
3, 4 or more unique structures). In some embodiments, at least 5%, at least
10%, at least 25%, at
least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic
acid is replaced with a
modified cytosine. The modified cytosine can be replaced by a compound having
a single unique
structure, or can be replaced by a plurality of compounds having different
structures (e.g., 2, 3, 4 or
more unique structures).
- 123 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Generally, the shortest length of a modified mRNA of the present disclosure
can be the
length of an mRNA sequence that is sufficient to encode for a dipeptide. In
another embodiment, the
length of the mRNA sequence is sufficient to encode for a tripeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a tetrapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a pentapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a hexapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a heptapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for an octapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a nonapeptide. In
another embodiment, the
length of an mRNA sequence is sufficient to encode for a decapeptide.
Examples of dipeptides that the modified nucleic acid sequences can encode for
include,
but are not limited to, carnosine and anserine.
In a further embodiment, the mRNA is greater than 30 nucleotides in length. In
another
embodiment, the RNA molecule is greater than 35 nucleotides in length. In
another embodiment, the
length is at least 40 nucleotides. In another embodiment, the length is at
least 45 nucleotides. In
another embodiment, the length is at least 55 nucleotides. In another
embodiment, the length is at
least 60 nucleotides. In another embodiment, the length is at least 60
nucleotides. In another
embodiment, the length is at least 80 nucleotides. In another embodiment, the
length is at least 90
nucleotides. In another embodiment, the length is at least 100 nucleotides. In
another embodiment,
the length is at least 120 nucleotides. In another embodiment, the length is
at least 140 nucleotides.
In another embodiment, the length is at least 160 nucleotides. In another
embodiment, the length is at
least 180 nucleotides. In another embodiment, the length is at least 200
nucleotides. In another
embodiment, the length is at least 250 nucleotides. In another embodiment, the
length is at least 300
nucleotides. In another embodiment, the length is at least 350 nucleotides. In
another embodiment,
the length is at least 400 nucleotides. In another embodiment, the length is
at least 450 nucleotides.
In another embodiment, the length is at least 500 nucleotides. In another
embodiment, the length is at
least 600 nucleotides. In another embodiment, the length is at least 700
nucleotides. In another
embodiment, the length is at least 800 nucleotides. In another embodiment, the
length is at least 900
nucleotides. In another embodiment, the length is at least 1000 nucleotides.
In another embodiment,
the length is at least 1100 nucleotides. In another embodiment, the length is
at least 1200
nucleotides. In another embodiment, the length is at least 1300 nucleotides.
In another embodiment,
the length is at least 1400 nucleotides. In another embodiment, the length is
at least 1500
nucleotides. In another embodiment, the length is at least 1600 nucleotides.
In another embodiment,
the length is at least 1800 nucleotides. In another embodiment, the length is
at least 2000
nucleotides. In another embodiment, the length is at least 2500 nucleotides.
In another embodiment,
the length is at least 3000 nucleotides. In another embodiment, the length is
at least 4000
nucleotides. In another embodiment, the length is at least 5000 nucleotides,
or greater than 5000
nucleotides.
For example, the modified nucleic acids described herein can be prepared using
methods
that are known to those skilled in the art of nucleic acid synthesis.
- 124 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, the present disclosure provides methods, e.g., enzymatic,
of
preparing a nucleic acid sequence comprising a nucleotide, wherein the nucleic
acid sequence
comprises a compound of Formula Xl-a:
X
_Y1PY2y3B
OR
y2 A
1-141¨P=X
ORcl
Xl-a
wherein:
the nucleotide has decreased binding affinity;
denotes an optional double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or -CRaRb- when denotes a single bond, or U is -CRa- when
denotes a double bond;
A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, or a
peptide
comprising 2 to 12 amino acids;
X is 0 or S;
each of Y1 is independently selected from 0, -NRal, and SR;
each of Y2 and Y3 are independently selected from 0, -CRaRb-, NRc, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai is H;
_OR cl is OH at a pH of about 1 or ¨ORcl is 0- at physiological pH; and
B is nucleobase;
provided that the ring encompassing the variables A, B, U, Y2 and Y3 cannot be
ribose
The method comprising reacting a compound of Formula XIII:
/X \
yl ______ _y2 3 m.
6Rci U D
n \
Y1 A
XIII
b1
l
l, _NRaR5
each of Y1 is independently selected from _oRa
and ¨SRal, wherein Rai and
Rbl are each independently H or a counterion
with an RNA polymerase, and a cDNA template.
In some embodiments, the reaction is repeated from 1 to about 7,000 times.
- 125 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, B is a nucleobase of Formula XII-a, XII-b, or XII-c:
R2 0 R2
R1V 'N
, R4 R3,N)-NH R3,N
'-
-w-Lx 0
XII-a XII-b XII-c
wherein:
denotes a single or double bond;
X is 0 or S;
U and W are each independently CH or N;
V is 0, S, C or N;
wherein when V is C then R1 is H, C1_6 alkyl, C2_6 alkenyl, C2_6 alkynyl,
halo, or ¨OW,
wherein C1_6 alkyl, C2_6 alkenyl, and C2_6 alkynyl are each optionally
substituted with ¨OH, -NRaRb, -
SH, -C(0)R, -C(0)OR, -NHC(0)Rc, or -NHC(0)0Rc;
and wherein when V is 0, S, or N then R1 is absent;
R2 is H, -SRc, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are
attached
can form a 5- or 6-membered ring optionally substituted with 1-4 substituents
selected from halo, -
OH, -SH, -NRaRb, C1_20 alkyl, C2_20 alkenyl, C2_20 alkynyl, C1_20 alkoxy, or
C1_20 thioalkyl;
R3 is H or C1_20 alkyl;
R4 is H or C1_20 alkyl; wherein when denotes a double bond then R4 is
absent, or N-R4,
taken together, forms a positively charged N substituted with C1_20 alkyl;
Ra and Rb are each independently H, C1_20 alkyl, C2_20 alkenyl, C2_20 alkynyl,
or C6_20 aryl;
and
Rc is H, C1_20 alkyl, C2_20 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula XII-al , XII-a2, XII-a3, XII-
a4, or XII-
a5:
X R2 R2 R2 0
R1 RyL
NN,R4 R1 N NH
X I
N
'0 X 'N 0
XII-a XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the methods further comprise a nucleotide selected from
the group
consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In another aspect, the present disclosure provides for methods of amplifying a
nucleic acid
sequence, the method comprising:
reacting a compound of Formula Xl-d:
- 126 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
X X X
vi_p y2 _ _y2 B
'
ORcl ORcl ORcl z
Y1 A
Xl-d
wherein:
denotes a single or a double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or -CRaRb- when
denotes a single bond, or U is -CRa- when
denotes a double bond;
Z is H, C1_12 alkyl, or C6_20 aryl, or Z is absent when denotes a double
bond; or
Z is -CRaRb- and forms a bond with A;
A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, or a
peptide
comprising 1 to 12 amino acids;
X is 0 or S;
bl, l
l, _NRaR
each of Y1 is independently selected from _oRa and ¨SRal;
each of Y2 and Y3 is independently selected from 0, -CRaRb-, NRc, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
B is nucleobase;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai and Rbi are each independently H or a counterion; and
_OR cl is OH at a pH of about 1 or ¨ORcl is 0- at physiological pH;
provided that the ring encompassing the variables A, B, U, Z, Y2 and Y3 cannot
be ribose
with a primer, a cDNA template, and an RNA polymerase.
In some embodiments, B is a nucleobase of Formula XII-a, XII-b, or XII-c:
R2 0 R2
R1, R4 R3, R3,
V N NH N N
,L
w x 0
XII-a XII-b XII-c
wherein:
denotes a single or double bond;
X is 0 or S;
U and W are each independently CH or N;
V is 0, S, C or N;
- 127 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
wherein when V is C then R1 is H, C1_6 alkyl, C2_6 alkenyl, C2_6 alkynyl,
halo, or ¨OW,
wherein C1_6 alkyl, C2_6 alkenyl, and C2_6 alkynyl are each optionally
substituted with ¨OH, -NRaRb, -
SH, -C(0)R, -C(0)OR, -NHC(0)Rc, or -NHC(0)0Rc;
and wherein when V is 0, S, or N then R1 is absent;
R2 is H, -SRc, -NRaRb, or halo;
or when V is C then R1 and R2 together with the carbon atoms to which they are
attached
can form a 5- or 6-membered ring optionally substituted with 1-4 substituents
selected from halo, -
OH, -SH, -NRaRb, C1_20 alkyl, C2_20 alkenyl, C2_20 alkynyl, C1_20 alkoxy, or
C1_20 thioalkyl;
R3 is H or C1_20 alkyl;
R4 is H or C1_20 alkyl; wherein when denotes a double bond then R4 is
absent, or
taken together, forms a positively charged N substituted with C1_20 alkyl;
Ra and RID are each independently H, C1_20 alkyl, C2_20 alkenyl, C2_20
alkynyl, or C6_20 aryl;
and
Rc is H, C1_20 alkyl, C2_20 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group.
In some embodiments, B is a nucleobase of Formula XII-al XII-a2, XII-a3, XII-
a4, or XII-
a5:
X R2 R2 R2 0
RN R4 RyLNH
NR4 " NN,R4 N R
XNX
0 -N 0 NO
XII-al XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the methods further comprise a nucleotide selected from
the group
consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In some embodiments, the present disclosure provides for methods of
synthesizing a
pharmaceutical nucleic acid, comprising the steps of:
a) providing a complementary deoxyribonucleic acid (cDNA) that encodes a
pharmaceutical protein of interest;
b) selecting a nucleotide and
c) contacting the provided cDNA and the selected nucleotide with an RNA
polymerase,
under conditions such that the pharmaceutical nucleic acid is synthesized.
In further embodiments, the pharmaceutical nucleic acid is a ribonucleic acid
(RNA).
In still a further aspect of the present disclosure, the modified nucleic
acids can be
prepared using solid phase synthesis methods.
- 128 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, the present disclosure provides methods of synthesizing a
nucleic
acid comprising a compound of Formula Xl-a:
X
B
6Rci
y2 A
1-141¨P=X
ORcl
Xl-a
wherein:
denotes an optional double bond;
- - -denotes an optional single bond;
U is 0, S, -NRa-, or -CRaRb- when denotes a single bond, or U is -CRa-
when
denotes a double bond;
A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, or a
peptide
comprising 2 to 12 amino acids;
X is 0 or S;
each of Y1 is independently selected from 0, NRal, and S;
each of Y2 and Y3 are independently selected from 0, -CRaRb-, NRc, S or a
linker
comprising one or more atoms selected from the group consisting of C, 0, N,
and S;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group;
Rai is H;
_OR cl is OH at a pH of about 1 or ¨ORcl is 0- at physiological pH; and
B is nucleobase;
provided that the ring encompassing the variables A, B, U, Y2 and Y3 cannot be
ribose;
comprising:
a) reacting a nucleotide of Formula XIII-a:
ylL y3
B
cry2 A 'F:s2
XIII-a
b1
l
l, _NRaR5
wherein each of Y1 is independently selected from _oRa
and ¨SRal, wherein
Rai and Rb1 are each independently H or a counterion;
- 129 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
with a phosphoramidite compound of Formula XIII-b:
p1 _y2_
Y3 U B
v2 A¨P2
P\3 1
0¨P\ (
XXIII-b
wherein: Ci) denotes a solid support; and
P1, P2 and P3 are each independently suitable protecting groups;
to provide a nucleic acid of Formula XIV-a:
p1 _ y2
vi2 A¨P2
P\3
0¨P
y1__ y3 u B
A¨P2
0y2
XIV-a and b) oxidizing or sulfurizing the nucleic acid of Formula XIV-a to
yield a nucleic acid of
Formula XlVb:
p1 _ y2 _ y3
A¨P2
P\3
y2 _ y3 u
2
A¨P2
(YY
XIV-b
and c) removing the protecting groups to yield the nucleic acid of Formula Xl-
a.
In some embodiments, the methods further comprise a nucleotide selected from
the group
consisting of adenosine, cytosine, guanosine, and uracil.
- 130 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In some embodiments, B is a nucleobase of Formula XIII:
R3
R44
XIII
wherein:
V is N or positively charged NRc;
R3 is NRcRd, -0Ra, or -SRa;
R4 is H or forms a bond with Y3;
R5 is H, -NRcRd, or -0Ra;
Ra and Rb are each independently H, C1_12 alkyl, C2_12 alkenyl, C2_12 alkynyl,
or C6_20 aryl;
and
Rc is H, C1_12 alkyl, C2_12 alkenyl, phenyl, benzyl, a polyethylene glycol
group, or an amino-
polyethylene glycol group.
In some embodiments, steps a) and b) are repeated from 1 to about 10,000
times.
5' Capping
The 5' cap structure of an mRNA is involved in nuclear export, increasing mRNA
stability and
binds the mRNA Cap Binding Protein (CBP), which is responsible for mRNA
stability in the cell and
translation competency through the association of CBP with poly(A) binding
protein to form the mature
cyclic mRNA species. The cap further assists the removal of 5' proximal
introns removal during mRNA
splicing.
Endogenous mRNA molecules may be 5'-end capped generating a 5'-ppp-5'-
triphosphate
linkage between a terminal guanosine cap residue and the 5'-terminal
transcribed sense nucleotide of the
mRNA. This 5'-guanylate cap may then be methylated to generate an N7-methyl-
guanylate residue. The
ribose sugars of the terminal and/or anteterminal transcribed nucleotides of
the 5' end of the mRNA may
optionally also be 2'-0-methylated. 5'-decapping through hydrolysis and
cleavage of the guanylate cap
structure may target a nucleic acid molecule, such as an mRNA molecule, for
degradation.
Modifications to the nucleic acids of the present invention may generate a non-
hydrolyzable
cap structure preventing decapping and thus increasing mRNA half-life. Because
cap structure
hydrolysis requires cleavage of 5'-ppp-5' phosphorodiester linkages, modified
nucleotides may be used
during the capping reaction. For example, a Vaccinia Capping Enzyme from New
England Biolabs
(Ipswich, MA) may be used with a-thio-guanosine nucleotides according to the
manufacturer's
instructions to create a phosphorothioate linkage in the 5'-ppp-5' cap.
Additional modified guanosine
nucleotides may be used such as a-methyl-phosphonate and seleno-phosphate
nucleotides.
Additional modifications include, but are not limited to, 2'-0-methylation of
the ribose sugars of
5'-terminal and/or 5'-anteterminal nucleotides of the mRNA (as mentioned
above) on the 2'-hydroxyl
group of the sugar ring. Multiple distinct 5'-cap structures can be used to
generate the 5'-cap of a nucleic
acid molecule, such as an mRNA molecule.
-131 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
5' Cap structures include those described in International Patent Publication
Nos.
W02008127688, WO 2008016473, and WO 2011015347, the 5' cap structures of which
are incorporated
herein by reference in its entirety.
Cap analogs, which herein are also referred to as synthetic cap analogs,
chemical caps,
chemical cap analogs, or structural or functional cap analogs, differ from
natural (i.e. endogenous, wild-
type or physiological) 5'-caps in their chemical structure, while retaining
cap function. Cap analogs may
be chemically (i.e. non-enzymatically) or enzymatically synthesized and/linked
to a nucleic acid molecule.
For example, the Anti-Reverse Cap Analog (ARCA) cap contains two guanines
linked by a 5'-
5'-triphosphate group, wherein one guanine contains an N7 methyl group as well
as a 3'-0-methyl group
(i.e., N7,3'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine (m7G-3'mppp-G;
which may equivalently
be designated 3' 0-Me-m7G(5')ppp(5')G). The 3'-0 atom of the other,
unmodified, guanine becomes
linked to the 5'-terminal nucleotide of the capped nucleic acid molecule (e.g.
an mRNA or mmRNA). The
N7- and 3'-0-methlyated guanine provides the terminal moiety of the capped
nucleic acid molecule (e.g.
mRNA or mmRNA).
Another exemplary cap is mCAP, which is similar to ARCA but has a 2'-0-methyl
group on
guanosine (i.e., N7,2'-0-dimethyl-guanosine-5'-triphosphate-5'-guanosine, m7Gm-
ppp-G).
In one embodiment, the cap is a dinucleotide cap analog. As a non-limiting
example, the
dinucleotide cap analog may be modified at different phosphate positions with
a boranophosphate group
or a phophoroselenoate group such as the dinucleotide cap analogs described in
US Patent No.
8,519,110, the caps of which are herein incorporated by reference in its
entirety.
In another embodiment, the cap analog is a N7-(4-chlorophenoxyethyl)
substituted
dicnucleotide form of a cap analog known in the art and/or described herein.
Non-limiting examples of a
N7-(4-chlorophenoxyethyl) substituted dinucleotide form of a cap analog
include a N7-(4-
chlorophenoxyethyl)-G(5)ppp(5')G and a N7-(4-chlorophenoxyethyl)-m3'-
G(5)ppp(5')G cap analog (See
e.g., the various cap analogs and the methods of synthesizing cap analogs
described in Kore et al.
Bioorganic & Medicinal Chemistry 2013 21:4570-4574; the caps of which are
herein incorporated by
reference in its entirety). In another embodiment, a cap analog of the present
invention is a 4-
chloro/bromophenoxyethyl analog.
While cap analogs allow for the concomitant capping of a nucleic acid molecule
in an in vitro
transcription reaction, up to 20% of transcripts remain uncapped. This, as
well as the structural
differences of a cap analog from endogenous 5'-cap structures of nucleic acids
produced by the
endogenous, cellular transcription machinery, may lead to reduced
translational competency and reduced
cellular stability.
Modified nucleic acids of the invention may also be capped post-
transcriptionally, using
enzymes, in order to generate more authentic 5'-cap structures. As used
herein, the phrase "more
authentic" refers to a feature that closely mirrors or mimics, either
structurally or functionally, an
endogenous or wild type feature. That is, a "more authentic" feature is better
representative of an
endogenous, wild-type, natural or physiological cellular function and/or
structure as compared to synthetic
features or analogs, etc., of the prior art, or which outperforms the
corresponding endogenous, wild-type,
natural or physiological feature in one or more respects. Non-limiting
examples of more authentic 5'-cap
structures of the present invention are those which, among other things, have
enhanced binding of cap
- 132 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
binding proteins, increased half life, reduced susceptibility to 5'
endonucleases and/or reduced 5'
decapping, as compared to synthetic 5'-cap structures known in the art (or to
a wild-type, natural or
physiological 5'-cap structure). For example, recombinant Vaccinia Virus
Capping Enzyme and
recombinant 2'-0-methyltransferase enzyme can create a canonical 5'-5'-
triphosphate linkage between
the 5'-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the
cap guanine contains an
N7 methylation and the 5'-terminal nucleotide of the mRNA contains a 2'-0-
methyl. Such a structure is
termed the Cap1 structure. This cap results in a higher translational-
competency and cellular stability and
a reduced activation of cellular pro-inflammatory cytokines, as compared,
e.g., to other 5'cap analog
structures known in the art. Cap structures include 7mG(5')ppp(5')N,pN2p (cap
0),
7mG(5')ppp(5')NImpNp (cap 1), 7mG(5')-ppp(5')NImpN2mp (cap 2) and
m(7)Gpppm(3)(6,6,2')Apm(2')Apm(2')Cpm(2)(3,2')Up (cap 4).
Because the modified nucleic acids may be capped post-transcriptionally, and
because this
process is more efficient, nearly 100% of the modified nucleic acids may be
capped. This is in contrast to
¨80% when a cap analog is linked to an mRNA in the course of an in vitro
transcription reaction.
According to the present invention, 5' terminal caps may include endogenous
caps or cap
analogs. According to the present invention, a 5' terminal cap may comprise a
guanine analog. Useful
guanine analogs include inosine, N1-methyl-guanosine, 211uoro-guanosine, 7-
deaza-guanosine, 8-oxo-
guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
In one embodiment, the nucleic acids described herein may contain a modified
5'-cap. A
modification on the 5'-cap may increase the stability of mRNA, increase the
half-life of the mRNA, and
could increase the mRNA translational efficiency. The modified 5'-cap may
include, but is not limited to,
one or more of the following modifications: modification at the 2' and/or 3'
position of a capped guanosine
triphosphate (GTP), a replacement of the sugar ring oxygen (that produced the
carbocyclic ring) with a
methylene moiety (CH2), a modification at the triphosphate bridge moiety of
the cap structure, or a
modification at the nucleobase (G) moiety.
The 5'-cap structure that may be modified includes, but is not limited to, the
caps described
herein such as Cap having the substrate structure for cap dependent
translation of:
9H OH NH
9 9o
0-P-O-P-0-17=-0-1 0 I
OH OH OH
H2NH,Try
OH
mRNA
0
(CAP-001), or Cap1 having the substrate
0
OH 9H e A.
- o N NH2
I -4)- 0
H2N N Cori om 61-1
Y OMe
HN,
/r CH mRNA
structure for cap dependent translation of: 0 CH2
(CAP-002).
- 133 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
As a non-limiting example, the modified 5'-cap may have the substrate
structure for cap
dependent translation of:
0
/,I4 114
0 9
\
0 0 0 N P. .k. NH2
II II
f
o-A-o-A--o-A-o--, ,
H2N N N OH 61-1 6H j
0 OH
6 ¨?zsClia ,
mRNA
(CAP-003),
0
N
--'ki--
0 0 )Ltal
N N NH2
I
i7 0 0 'IT 0--_c_))
OH OH OH
/42N, N N
-r-..- -IT- \
HP, 1 /1 0 011-1
N mRNA
6 ' µCE-13 (CAP-004),
0
Mi-kw
OMe OH <
N [ ININH2
1
----1 -0- 11-0I-010 0 /
I icr4
H2N,y,, 011 OH OH
,
Hr4 I 4> 9 OH
EP\CH mRNA
3
0 (CAP-005),
0
OH 9Me N 1k
0 0 0 1:i Pi ' Ni-12
.....Ø-o-ii-o-1:1-o-il-o 0 '
H2N,,.INT,I.P1.). 011 0H
HN 1 / 0 OH
N
0 : ETRNA
0 CH3 (CAP-006),
9
Nxii- NH
OMe Owe: I ,
0 0 0 N N NH2
14.-- ---0-4-0-P--0-1,L0 ..,0
, OH OH 0H
H211 N N
111jN 0 OH
õ., mRNA
0 (CAP-007),
0
Ni-J1-- NH
0 0 0 N
NH2 -"-
L-04-0-P-0-P-0 0 1
H2Plyp - 6H 611 6H )--- --?1
HP 0 OH
mRNA
0 cils (CAP-008),
Q N 0
0 0 j(l'iFi
-1:4-0-1)L60-2L-0 0 N N-A's NH2
OH OH OH -_4
--- ,,>
9 OH
mRNA
6 8 ' ciis (CAP-009),
- 134 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
n N 0
r0 0
0 0 0
H N NH2
1
(t 3L04 0 I' 0 Vo---
H 1 0 I
OH 011 Om
HN .......õ
ic....cltii
0 OH
,
mRNA
19 bil, (CAP-010),
H . 0
NA
0 0 0
LI N I N,I NH2
1
C)- -0-0-0 P 0-1,1-0 0õ..
H2N,1õ,...NN\ OH OH OH
' II //' ? OH
mRNA
I . '
0 CH3
(CAP-011),
x..0,,, He
H
0
N
111'Z
0 0
N
0 0 0 N NH2
I
--0---0-11-0-P-0-11-0 0
i N N N
H 694 614
64
2 Irti.µ 0 OH
PIN
mRNA
O
S =CH3
(CAP-012),
o
?fi !4-1"(NH
0-.......L
N N NH2
0 _01-0-17-01-0- 0 1
'---''
H2N
OH OH OH c......
:1,14
HN' I N ? OH
mRNA
8 '=3 'as
(CAP-013),
o
N
OH OH <,, 111111:1
i
N Pr NH2 0 0 0 1
1-0-4-0.-14 0 P0 - 0
H2N ON OH 0H .......
,T.;:iirIN
0 OH
HN I
mRNA
o
eCH3
(CAP-014),
0
N
1111*--A4H,
00
N N NH2
1
-0 4-0-0-04-07 0 1
I
H2N,,,..N.õ.N OH Om OH
1 , =,.?
HN 0 OH,
---N
MRNA
il 0 =
o CH3
(CAP-015),
- 135 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
9
OH OH
4-1-- S 0 0 til N''NH2
- 0 -P-04-0-114-0-1c:_?3õ 1
H2N N N OH OH OH
---,--:-- --rr-- \
Ht1.1=.µ 0 OH
mRNA
OH µ 1.13 (CAP-016),
0
N, ii,1,,,IH
OH OH ..
.. .. k.,
00 0 .,.. : . NH2
, t:71k ii 1, ii i
0 -P-O-P- 0-P-0
i.vo ...),- A SH3 011 OH
8
:=,) 9 9"
= -14 mRNA
(CAP-017),
0
OH OH <,./t4 ii)11:4 ii
41-111_ 0 0 0 N lek- NH2
1 0 -0-P-04-04-0--, 0 !
H2Nis, .. N CH3 OH OH
u.., N> 0 OH
, = mRNA
(CAP-018),
0
N,õõ..1-.. NH
OH OH =1, .1.,
Si 9 9 r.._ . N NI-12
i
621-4 OH OH
H2N, =
9 OH
1s-
i , . )
mRNA
0 CH3 (CAP-01 9),
0
14------- NH
OH OH 1 ,-(=
4--- 0 0 N"1.111C11 NH2
!.-
H2NyN 6cH3 OH OH
y14\
0 OH
mRNA
6 cH.,. (CAP-020),
0[1..
OH OH /31..õ NH
\ 4
S 9 9 1,:i.}..N1-1,4--,Nti2 ¨Fik
1
:,--0--04
H2N N` N OCH3 C.- 0
'---(:- y" \.,).
Hil.õ_2 .,. 0 OH
n El mRNA
6 CH3 (CAP-021),
0
N.
1,
OR, OR2
-
0 0 0
0- 41,- 1 -A-0-41.0- 0 N.11''' : . 'NH2
1.----
OH OH OH
H2N N. N
Y T-- ' 9 0H
HN ).... I/
N
e, rreRNA
6 cis where R1 and R2 are defined
in Table 4:
Table 4: R1 and R2 groups for CAP-022 to CAP096.
- 136 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Cap R1 R2
Structure
Number
CAP-022 C2H5 (Ethyl) H
CAP-023 H C2H5 (Ethyl)
CAP-024 C2H5 (Ethyl) C2H5 (Ethyl)
CAP-025 C3H7 (Propyl) H
CAP-026 H C3H7 (Propyl)
CAP-027 C3H7 (Propyl) C3H7 (Propyl)
CAP-028 C4H9 (Butyl) H
CAP-029 H C4H9 (Butyl)
CAP-030 C4H9 (Butyl) C4H9 (Butyl)
CAP-031 C5H11 (Pentyl) H
CAP-032 H C5H11 (Pentyl)
CAP-033 C5H11 (Pentyl) C5H11 (Pentyl)
CAP-034 H2C-CECH (Propargyl) H
CAP-035 H H2C-CECH (Propargyl)
CAP-036 H2C-CECH (Propargyl) H2C-CECH (Propargyl)
CAP-037 CH2CH=CH2 (Ally!) H
CAP-038 H CH2CH=CH2 (Ally!)
CAP-039 CH2CH=CH2 (Ally!) CH2CH=CH2 (Ally!)
CAP-040 CH2OCH3 (MOM) H
CAP-041 H CH2OCH3 (MOM)
CAP-042 CH2OCH3 (MOM) CH2OCH3 (MOM)
CAP-043 CH2OCH2CH2OCH3 (MEM) H
CAP-044 H CH2OCH2CH2OCH3 (MEM)
CAP-045 CH2OCH2CH2OCH3 (MEM) CH2OCH2CH2OCH3 (MEM)
CAP-046 CH2SCH3(MTM) H
CAP-047 H CH2SCH3(MTM)
CAP-048 CH2SCH3(MTM) CH2SCH3(MTM)
CAP-049 CH2C6H5 (Benzyl) H
CAP-050 H CH2C6H5 (Benzyl)
CAP-051 CH2C6H5 (Benzyl) CH2C6H5 (Benzyl)
CAP-052 CH2OCH2C6H5 (BOM) H
CAP-053 H CH2OCH2C6H5 (BOM)
CAP-054 CH2OCH2C6H5 (BOM) CH2OCH2C6H5 (BOM)
CH2C6H4-0Me (p- H
CAP-055 Methoxybenzyl)
H CH2C6H4-0Me (p-
CAP-056 Methoxybenzyl)
CH2C6H4-0Me (p- CH2C6H4-0Me (p-
CAP-057 Methoxybenzyl) Methoxybenzyl)
CAP-058 CH2C6H4-NO2 (p-Nitrobenzyl) H
CAP-059 H CH2C6H4-NO2 (p-Nitrobenzyl)
CAP-060 CH2C6H4-NO2 (p-Nitrobenzyl) CH2C6H4-NO2 (p-Nitrobenzyl)
CH2C6H4-X (p-Halobenzyl) H
CAP-061 where X=F, Cl, Br or I
H CH2C6H4-X (p-Halobenzyl)
CAP-062 where X=F, Cl, Br or I
CH2C6H4-X (p-Halobenzyl) CH2C6H4-X (p-Halobenzyl)
CAP-063 where X=F, Cl, Br or I where X=F, Cl, Br or I
CAP-064 CH2C6H4-N3 (p-Azidobenzyl) H
CAP-065 H CH2C6H4-N3 (p-Azidobenzyl)
CAP-066 CH2C6H4-N3 (p-Azidobenzyl) CH2C6H4-N3 (p-Azidobenzyl)
CH2C61--14-CF3 (P- H
CAP-067 Trifluoromethylbenzyl)
H CH2C61--14-CF3 (P-
CAP-068 Trifluoromethylbenzyl)
- 137 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
CH2C6H4-CF3 (P- CH2C6H4-CF3 (P-
CAP-069 Trifluoromethylbenzyl) Trifluoromethylbenzyl)
CH2C6H4-0CF3 (p-
CAP-070 Trifluoromethoxylbenzyl)
CH2C6H4-0CF3 (p-
CAP-071 Trifluoromethoxylbenzyl)
CH2C6H4-0CF3 (p- CH2C6H4-0CF3 (p-
CAP-072 Trifluoromethoxylbenzyl) Trifluoromethoxylbenzyl)
CH2C6H3-(CF3)2 [2,4-
CAP-073 bis(Trifluoromethyl)benzyl]
CH2C6H3-(CF3)2 [2,4-
CAP-074 bis(Trifluoromethyl)benzyl]
CH2C6H3-(CF3)2 [2,4- CH2C6H3-(CF3)2 [2,4-
CAP-075 bis(Trifluoromethyl)benzyl] bis(Trifluoromethyl)benzyl]
Si(C6H5)2C4H9 (t-
CAP-076 Butyldiphenylsily1)
Si(C6H5)2C4H9 (t-
CAP-077 Butyldiphenylsily1)
Si(C6H5)2C4H9 (t- Si(C6H5)2C4H9 (t-
CAP-078 Butyldiphenylsily1) Butyldiphenylsily1)
CAP-079 CH2CH2CH=CH2 (Homoally1)
CAP-080 H CH2CH2CH=CH2 (Homoally1)
CAP-081 CH2CH2CH=CH2 (Homoally1) CH2CH2CH=CH2 (Homoally1)
CAP-082 P(0)(OH)2 (MP)
CAP-083 H P(0)(01-1)2 (MP)
CAP-084 P(0)(OH)2 (MP) P(0)(01-1)2 (MP)
CAP-085 P(S)(OH)2 (Thio-MP)
CAP-086 H P(S)(OH)2 (Thio-MP)
CAP-087 P(S)(OH)2 (Thio-MP) P(S)(OH)2 (Thio-MP)
P(0)(CH3)(OH)
CAP-088 (Methylphophonate)
P(0)(CH3)(OH)
CAP-089 (Methylphophonate)
P(0)(CH3)(OH) P(0)(CH3)(OH)
CAP-090 (Methylphophonate) (Methylphophonate)
PN(Pr)2(OCH2CH2CN)
CAP-091 (Phosporamidite)
PN(Pr)2(OCH2CH2CN)
CAP-092 (Phosporamidite)
PN(Pr)2(0CH2CH2CN) PN(Pr)2(0CH2CH2CN)
CAP-093 (Phosporamidite) (Phosporamidite)
CAP-094 SO2CH3 (Methanesulfonic acid) H
CAP-095 H SO2CH3 (Methanesulfonic acid)
CAP-096 SO2CH3 (Methanesulfonic acid) SO2CH3 (Methanesulfonic acid)
0
Ri R2
N
0 0 H2
H2I" OH OH OH
ii 9 OH
InRNA
or b113 where R1 and R2 are defined in Table
5:
- 138 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 5: R1 and R2 groups for CAP-097 to CAP111.
Cap Structure R1 R2
Nurnber
CAP-097 NH2 (amino) H
CAP-098 H NH2 (amino)
CAP-099 NH2 (amino) NH2 (amino)
CAP-100 N3 (Azido) H
CAP-101 H N3 (Azido)
CAP-102 N3 (Azido) N3 (Azido)
CAP-103 X (Halo: F, Cl, Br, I) H
CAP-104 H X (Halo: F, Cl, Br, I)
CAP-105 X (Halo: F, Cl, Br, I) X (Halo: F, Cl, Br, I)
CAP-106 SH (Thiol) H
CAP-107 H SH (Thiol)
CAP-108 SH (Thiol) SH (Thiol)
CAP-109 SCH3 (Thiomethyl) H
CAP-110 H SCH3 (Thiomethyl)
CAP-111 SCH3 (Thiomethyl) SCH3 (Thiomethyl)
In Table 4, "MOM" stands for methoxymethyl, "MEM" stands for
methoxyethoxymethyl, "MTM"
stands for methylthiomethyl, "BOM" stands for benzyloxymethyl and "MP" stands
for monophosphonate.
In Table 4 and 5, "F" stands for fluorine, "Cl" stands for chlorine, "Br"
stands for bromine and "I" stands for
iodine.
In a non-limiting example, the modified 5'cap may have the substrate structure
for vaccinia
mRNA capping enzyme of:
0 o
i,N"----A NH I}
\N.A'N--)'"N112 c
NH
9 aN-,N142
0 , . o o o
Ho-fo-A- , .0 0 HO- r. i,
P-0 P-O-P-0-- 0
OH OH 7H - 6H OH OH c....._.
oxo
-6.-X L
(CAP-112),
(CAP-113),
o o
N----'1'NH N----)is'NH
0 0 0 N N NH 2 ? 2 NH
ii ii ii
HO-P-O-P-O-P-0- HO--Q4-O--O-10
r
6H 6H 6H OH 6H 611
OH Oalle (CAP-114), OMe OH (CAP-115),
0
o
NH
N .NH2
NA...-..%1,.l
0 0 0 0 0 0
II ii ii II II ii
HO--O--O---OT c ,N NH2 0
6H 01-1 OH 6H 6H 6ii
Wile OMe (CAP-116), o o
-.....- (CAP-
117),
- 139 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
o
0
N---ANH
I :
iiii
N.x11--
<,, Nit
0 0 0
? 0 0 N ers'NH2
H04- f:1` 4 .':.' A-0----1 0
.-i.:4 -o-p---o?
611 6H OH
Orx?
6.xO
S-, __ 7 (CAP-118), ",---- (CAP-119),
o 0
N....TANH
-1: ...r ,.
0 0 0 ,..1.- = ,41-12 0 0 0 N N NH2
I
il II II
FICI-A-04-044-0 0
HO--C -F-0--p--07
OH OH OH --- OH OH OH
00 _ ,,,0 0
PH (CAP-120), H
Me0.'-'"''T. --...
(CAP-121),
0
0
N
YIE1 N
0 0
NA'NNH2
1
0
N N NH2
II II tl r 0 0 0
HO-P-O-P-0-!-O¨ _0 i
1
OH OH OH '-",c.... H04-04-0-0-0
6H OH OH -1Spi
'-i __ 0
i
OH (CAP-122), OH OH (CAP-123),
0
9
N
Isl...,
c4).õN,,NH2 71
N 1µ1.- NH2
0 0 0 0 0 s
HO-P-O-P-O4-07 I ii ii II
HO-12-0-P-0-P-0-0
OH OH OH kc:7---
OH OH OH
00
A (CAP-124), OH OH (CAP-125),
o 0
N
9 9 9 N N NH2
-11:17.
N,,t NNI.12
0 0 0
HO-p-0-1?-0-P-00 HO-P-0 -iL0-4- 0 0
OH OH BH3 ________________
8 - CSH OH 0E13
OH OH
(CAP-126), OH OH (CAP-127),
0 o
N-----A NH FL....)1-.N H
N,,t NNH2NNNH2
6 0 0 0 0 0
!I r ii II II II
HO-P-O-P-0-1-00 HO-P-O-P-O-P-0 0
6H 6 H 02H5 61-I 6 H 6cH3
(5H OH (CAP-128), OH OH (CAP-
129),
o
o
N---)1 N14H ljtri
NH2
0 0 S 0 0 0 N N NH2
II II B
II II El
HO-P-O-P-O-P-0 0 HO-P-O-P-O-P-
C)
I I 1
I I I
OH OH OCH3 OH OH OF -----?
OH OH (CAP-130), OH OCH3 (CAP-
131),
- 140 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
0
NjN
(x.
N N NH 0 0 0 N N NH2
H01-01-0-9P-0 0 HO-12.-04-0-13-0 0
OH OH OH OH OH OH
[
OH F (CAP-132), OH N3 (CAP-133),
9 9 0 res'N NH2 o o
II ii II II NH
HO-P-0-13-0-1O 0 HO-P-O-P-O-P-0
OH OH OH OH OH
OH NH2 (CAP-134), OH OH
(CAP-135),
Ni*NH
I
0 0 0 NH2
HO-P-O-P-04-0 0
OH OH OH
OR2 ORi where R1 and R2 are defined in Table 6:
Table 6: R1 and R2 groups for CAP-136 to CAP-210.
Cap R1 R2
Structure
Nurnber
CAP-136 C2H5 (Ethyl)
CAP-137 H C2H5 (Ethyl)
CAP-138 C2H5 (Ethyl) C2H5 (Ethyl)
CAP-139 C3H7 (Propyl)
CAP-140 H C3H7 (Propyl)
CAP-141 C3H7 (Propyl) C3H7 (Propyl)
CAP-142 C4H9 (Butyl)
CAP-143 H C4H9 (Butyl)
CAP-144 C4H9 (Butyl) C4H9 (Butyl)
CAP-145 C5H11 (Pentyl)
CAP-146 H C5H11 (Pentyl)
CAP-147 C5H11 (Pentyl) C5H11 (Pentyl)
CAP-148 H2C-CECH (Propargyl)
CAP-149 H H2C-CECH (Propargyl)
CAP-150 H2C-CECH (Propargyl) H2C-CECH (Propargyl)
CAP-151 CH2CH=CH2 (Ally!)
CAP-152 H CH2CH=CH2 (Ally!)
CAP-153 CH2CH=CH2 (Ally!) CH2CH=CH2 (Ally!)
CAP-154 CH2OCH3 (MOM)
CAP-155 H CH2OCH3 (MOM)
CAP-156 CH2OCH3 (MOM) CH2OCH3 (MOM)
CAP-157 CH2OCH2CH2OCH3 (MEM)
CAP-158 H CH2OCH2CH2OCH3 (MEM)
CAP-159 CH2OCH2CH2OCH3 (MEM) CH2OCH2CH2OCH3 (MEM)
CAP-160 CH2SCH3(MTM)
CAP-161 H CH2SCH3(MTM)
CAP-162 CH2SCH3(MTM) CH2SCH3(MTM)
CAP-163 CH2C6H5 (Benzyl)
CAP-164 H CH2C6H5 (Benzyl)
- 141 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
CAP-165 CH2C6H5 (Benzyl) CH2C6H5 (Benzyl)
CAP-166 CH2OCH2C6H5 (BOM) H
CAP-167 H CH2OCH2C6H5 (BOM)
CAP-168 CH2OCH2C6H5 (BOM) CH2OCH2C6H5 (BOM)
CH2C6H4-0Me (p- H
CAP-169 Methoxybenzyl)
CAP-170 H CH2C6H4-0Me (p-Methoxybenzyl)
CH2C6H4-0Me (p- CH2C6H4-0Me (p-Methoxybenzyl)
CAP-171 Methoxybenzyl)
CH2C6H4-NO2 (P- H
CAP-172 Nitrobenzyl)
CAP-173 H CH2C6H4-NO2 (p-Nitrobenzyl)
CH2C6H4-NO2 (P- CH2C6H4-NO2 (p-Nitrobenzyl)
CAP-174 Nitrobenzyl)
CH2C6H4-X (p-Halobenzyl) H
CAP-175 where X=F, Cl, Broil
H CH2C6H4-X (p-Halobenzyl) where
CAP-176 X=F, Cl, Broil
CH2C6H4-X (p-Halobenzyl) CH2C6H4-X (p-Halobenzyl) where
CAP-177 where X=F, Cl, Broil X=F, Cl, Broil
CAP-178 CH2C6H4-N3 (p-Azidobenzyl) H
CAP-179 H CH2C6H4-N3 (p-Azidobenzyl)
CAP-180 CH2C6H4-N3 (p-Azidobenzyl) CH2C6H4-N3 (p-Azidobenzyl)
CH2C61--14-CP3 (P- H
CAP-181 Trifluoromethylbenzyl)
H CH2C61--14-CP3 (P-
CAP-182 Trifluoromethylbenzyl)
CH2C61--14-CP3 (P- CH2C61--14-CP3 (P-
CAP-183 Trifluoromethylbenzyl) Trifluoromethylbenzyl)
CH2C6H4-0CF3 (p- H
CAP-184 Trifluoromethoxylbenzyl)
H CH2C6H4-0CF3 (p-
CAP-185 Trifluoromethoxylbenzyl)
CH2C6H4-0CF3 (p- CH2C6H4-0CF3 (p-
CAP-186 Trifluoromethoxylbenzyl) Trifluoromethoxylbenzyl)
CH2C6H3-(CF3)2 [2,4- H
CAP-187 bis(Trifluoromethyl)benzyl]
H CH2C6H3-(CF3)2 [2,4-
CAP-188 bis(Trifluoromethyl)benzyl]
CH2C6H3-(CF3)2 [2,4- CH2C6H3-(CF3)2 [2,4-
CAP-189 bis(Trifluoromethyl)benzyl] bis(Trifluoromethyl)benzyl]
Si(C6H5)2C4H9 (t- H
CAP-190 Butyldiphenylsily1)
CAP-191 H Si(C6H5)2C4H9 (t-Butyldiphenylsily1)
Si(C6H5)2C4H9 (t- Si(C6H5)2C4H9 (t-Butyldiphenylsily1)
CAP-192 Butyldiphenylsily1)
CH2CH2CH=CH2 H
CAP-193 (Homoally1)
CAP-194 H CH2CH2CH=CH2 (Homoally1)
CH2CH2CH=CH2 CH2CH2CH=CH2 (Homoally1)
CAP-195 (Homoally1)
CAP-196 P(0)(OH)2 (MP) H
CAP-197 H P(0)(01-1)2 (MP)
CAP-198 P(0)(OH)2 (MP) P(0)(01-1)2 (MP)
CAP-199 P(S)(OH)2 (Thio-MP) H
CAP-200 H P(S)(OH)2 (Thio-MP)
CAP-201 P(S)(OH)2 (Thio-MP) P(S)(OH)2 (Thio-MP)
P(0)(CH3)(OH) H
CAP-202 (Methylphophonate)
- 142 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
CAP-203 H P(0)(CH3)(OH)
(Methylphophonate)
P(0)(CH3)(OH) P(0)(CH3)(OH)
(Methylphophonate)
CAP-204 (Methylphophonate)
PN(Pr)2(OCH2CH2CN)
CAP-205 (Phosporamidite)
PN(Pr)2(OCH2CH2CN)
CAP-206 (Phosporamidite)
PN(Pr)2(OCH2CH2CN) PN(Pr)2(OCH2CH2CN)
CAP-207 (Phosporamidite) (Phosporamidite)
SO2CH3 (Methanesulfonic
CAP-208 acid)
CAP-209 H SO2CH3 (Methanesulfonic acid)
SO2CH3 (Methanesulfonic SO2CH3 (Methanesulfonic acid)
CAP-210 acid)
N"--ANH
I
N
9 9 9 H2
HO--O--O-P-O 0
OH OH OH
or R2 R1 where R1 and R2 are defined in
Table 7:
Table 7: R1 and R2 groups for CAP-211 to 225.
Cap R1 R2
Structure
Nurnber
CAP-211 NH2 (amino)
CAP-212 H NH2 (amino)
CAP-213 NH2 (amino) NH2 (amino)
CAP-214 N3 (Azido)
CAP-215 H N3 (Azido)
CAP-216 N3 (Azido) N3 (Azido)
CAP-217 X (Halo: F, Cl, Br, I)
CAP-218 H X (Halo: F, Cl, Br, I)
CAP-219 X (Halo: F, Cl, Br, I) X (Halo: F, Cl, Br, I)
CAP-220 SH (Thiol)
CAP-221 H SH (Thiol)
CAP-222 SH (Thiol) SH (Thiol)
CAP-223 SCH3 (Thiomethyl)
CAP-224 H SCH3 (Thiomethyl)
CAP-225 SCH3 (Thiomethyl) SCH3 (Thiomethyl)
In Table 6, "MOM" stands for methoxymethyl, "MEM" stands for
methoxyethoxymethyl, "MTM"
stands for methylthiomethyl, "BOM" stands for benzyloxymethyl and "MP" stands
for monophosphonate.
In Table 6 and 7, "F" stands for fluorine, "Cl" stands for chlorine, "Br"
stands for bromine and "I" stands for
iodine.
In another non-limiting example, of the modified capping structure substrates
CAP-112 ¨ CAP-
225 could be added in the presence of vaccinia capping enzyme with a component
to create enzymatic
activity such as, but not limited to, S-adenosylmethionine (AdoMet), to form a
modified cap for mRNA.
In one embodiment, the replacement of the sugar ring oxygen (that produced the
carbocyclic
ring) with a methylene moiety (CH2) could create greater stability to the C-N
bond against phosphorylases
- 143 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
as the C-N bond is resitant to acid or enzymatic hydrolysis. The methylene
moiety may also increase the
stability of the triphosphate bridge moiety and thus increasing the stability
of the mRNA. As a non-limiting
example, the cap substrate structure for cap dependent translation may have
the structure such as, but
not limited to, CAP-014 and CAP-015 and/or the cap substrate structure for
vaccinia mRNA capping
enzyme such as, but not limited to, CAP-123 and CAP-124. In another example,
CAP-112 ¨ CAP-122
and/or CAP-125 ¨ CAP-225, can be modified by replacing the sugar ring oxygen
(that produced the
carbocyclic ring) with a methylene moiety (CH2)=
In another embodiment, the triphophosphate bridge may be modified by the
replacement of at
least one oxygen with sulfur (thio), a borane (BH3) moiety, a methyl group, an
ethyl group, a methoxy
group and/or combinations thereof. This modification could increase the
stability of the mRNA towards
decapping enzymes. As a non-limiting example, the cap substrate structure for
cap dependent translation
may have the structure such as, but not limited to, CAP-016 ¨ CAP-021 and/or
the cap substrate
structure for vaccinia mRNA capping enzyme such as, but not limited to, CAP-
125 ¨ CAP-130. In another
example, CAP-003 ¨ CAP-015, CAP-022 ¨ CAP-124 and/or CAP-131 ¨ CAP-225, can be
modified on the
triphosphate bridge by replacing at least one of the triphosphate bridge
oxygens with sulfur (thio), a
borane (BH3) moiety, a methyl group, an ethyl group, a methoxy group and/or
combinations thereof.
In one embodiment, CAP-001 ¨ 134 and/or CAP-136 ¨ CAP-225 may be modified to
be a
thioguanosine analog similar to CAP-135. The thioguanosine analog may comprise
additional
modifications such as, but not limited to, a modification at the triphosphate
moiety (e.g., thio, BH3, CH3,
C2H5, OCH3, S and S with OCH3), a modification at the 2' and/or 3' positions
of 6-thio guanosine as
described herein and/or a replacement of the sugar ring oxygen (that produced
the carbocyclic ring) as
described herein.
In one embodiment, CAP-001 ¨ 121 and/or CAP-123 ¨ CAP-225 may be modified to
be a
modified 5'cap similar to CAP-122. The modified 5'cap may comprise additional
modifications such as,
but not limited to, a modification at the triphosphate moiety (e.g., thio,
BH3, CH3, C2H5, OCH3, S and S
with OCH3), a modification at the 2' and/or 3' positions of 6-thio guanosine
as described herein and/or a
replacement of the sugar ring oxygen (that produced the carbocyclic ring) as
described herein.
In one embodiment, the 5'cap modification may be the attachment of biotin or
conjugation at
the 2' or 3' position of a GTP.
In another embodiment, the 5' cap modification may include a CF2 modified
triphosphate
moiety.
In another embodiment, the triphosphate bridge of any of the cap structures
described herein
may be replaced with a tetraphosphate or pentaphosphate bridge. Examples of
tetraphosphate and
pentaphosphate containing bridges and other cap modifications are described in
Jemielity, J. et al. RNA
2003 9:1108-1122; Grudzien-Nogalska, E. et al. Methods Mol. Biol. 2013 969:55-
72; and Grudzien, E. et
al. RNA, 2004 10:1479-1487, the cap modifications of which are incorporated
herein by reference in its
entirety.
Terminal Architecture Modifications: Stem Loop
In one embodiment, the nucleic acids of the present invention may include a
stem loop such
as, but not limited to, a histone stem loop. The stem loop may be a nucleotide
sequence that is about 25
- 144 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
or about 26 nucleotides in length such as, but not limited to, SEQ ID NOs: 7-
17 as described in
International Patent Publication No. W02013103659, the stem loop sequences of
which are incorporated
herein by reference. The histone stem loop may be located 3' relative to the
coding region (e.g., at the 3'
terminus of the coding region). As a non-limiting example, the stem loop may
be located at the 3' end of
a nucleic acid described herein.
In one embodiment, the stem loop may be located in the second terminal region.
As a non-
limiting example, the stem loop may be located within an untranslated region
(e.g., 3'UTR) in the second
terminal region.
In one embodiment, the nucleic acid such as, but not limited to mRNA, which
comprises the
histone stem loop may be stabilized by the addition of at least one chain
terminating nucleoside. Not
wishing to be bound by theory, the addition of at least one chain terminating
nucleoside may slow the
degradation of a nucleic acid and thus can increase the half-life of the
nucleic acid.
In one embodiment, the chain terminating nucleoside may be, but is not limited
to, those
described in International Patent Publication No. W02013103659, the chain
terminating nucleosides of
which are incorporated herein by reference. In another embodiment, the chain
terminating nucleosides
which may be used with the present invention includes, but is not limited to,
3'-deoxyadenosine
(cordycepin), 3'-deoxyuridine, 3'-deoxycytosine, 3'-deoxyguanosine, 3'-
deoxythymine, 2',3'-
dideoxynucleosides, such as 2',3'- dideoxyadenosine, 2',3'-dideoxyuridine,
2',3'-dideoxycytosine, 2',3'-
dideoxyguanosine, 2',3'-dideoxythymine, a 2'-deoxynucleoside, or a -0-
methylnucleoside.
In another embodiment, the nucleic acid such as, but not limited to mRNA,
which comprises
the histone stem loop may be stabilized by a modification to the 3'region of
the nucleic acid that can
prevent and/or inhibit the addition of oligio(U) (see e.g., International
Patent Publication No.
W02013103659, the modifications to the 3'region of which are incorporated
herein by reference).
In yet another embodiment, the nucleic acid such as, but not limited to mRNA,
which
comprises the histone stem loop may be stabilized by the addition of an
oligonucleotide that terminates in
a 3'-deoxynucleoside, 2',3'-dideoxynucleoside 3'-0- methylnucleosides, 3'-0-
ethylnucleosides, 3'-
arabinosides, and other modified nucleosides known in the art and/or described
herein.
In one embodiment, the nucleic acids of the present invention may include a
histone stem
loop, a polyA tail sequence and/or a 5'cap structure. The histone stem loop
may be before and/or after
the polyA tail sequence. The nucleic acids comprising the histone stem loop
and a polyA tail sequence
may include a chain terminating nucleoside described herein.
In another embodiment, the nucleic acids of the present invention may include
a histone stem
loop and a 5'cap structure. The 5'cap structure may include, but is not
limited to, those described herein
and/or known in the art.
In one embodiment, the conserved stem loop region may comprise a miR sequence
described
herein. As a non-limiting example, the stem loop region may comprise the seed
sequence of a miR
sequence described herein. In another non-limiting example, the stem loop
region may comprise a miR-
122 seed sequence.
In another embodiment, the conserved stem loop region may comprise a miR
sequence
described herein and may also include a TEE sequence.
- 145 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In one embodiment, the incorporation of a miR sequence and/or a TEE sequence
changes the
shape of the stem loop region which may increase and/or decrease translation.
(see e.g, Kedde et al. A
Pumilio-induced RNA structure switch in p27-3'UTR controls miR-221 and miR-22
accessibility. Nature
Cell Biology. 2010, the miR sequences and/or a TEE sequences of which are
incorporated by reference).
In one embodiment, the modified nucleic acids described herein may comprise at
least one
histone stem-loop and a polyA sequence or polyadenylation signal. Non-limiting
examples of nucleic acid
sequences encoding for at least one histone stem-loop and a polyA sequence or
a polyadenylation signal
are described in International Patent Publication No. W02013120497,
W02013120629, W02013120500,
W02013120627, W02013120498, W02013120626, W02013120499 and W02013120628, the
sequences of which are incorporated herein by reference in their entirety. In
one embodiment, the
nucleic acid encoding for a histone stem loop and a polyA sequence or a
polyadenylation signal may
code for a pathogen antigen or fragment thereof such as the nucleic acid
sequences described in
International Patent Publication No W02013120499 and W02013120628, the
sequences of both of
which are incorporated herein by reference in their entirety. In another
embodiment, the nucleic acid
encoding for a histone stem loop and a polyA sequence or a polyadenylation
signal may code for a
therapeutic protein such as the nucleic acid sequences described in
International Patent Publication No
W02013120497 and W02013120629, the sequences of both of which are incorporated
herein by
reference in their entirety. In one embodiment, the nucleic acid encoding for
a histone stem loop and a
polyA sequence or a polyadenylation signal may code for a tumor antigen or
fragment thereof such as the
nucleic acid sequences described in International Patent Publication No
W02013120500 and
W02013120627, the sequences of both of which are incorporated herein by
reference in their entirety. In
another embodiment, the nucleic acid encoding for a histone stem loop and a
polyA sequence or a
polyadenylation signal may code for a allergenic antigen or an autoimmune self-
antigen such as the
nucleic acid sequences described in International Patent Publication No
W02013120498 and
W02013120626, the sequences of both of which are incorporated herein by
reference in their entirety.
Terminal Architecture Modifications: 3'UTR and Triple Helices
In one embodiment, nucleic acids of the present invention may include a triple
helix on the 3'
end of the modified nucleic acid, enhanced modified RNA or ribonucleic acid.
The 3' end of the nucleic
acids of the present invention may include a triple helix alone or in
combination with a Poly-A tail.
In one embodiment, the nucleic acid of the present invention may comprise at
least a first and
a second U-rich region, a conserved stem loop region between the first and
second region and an A-rich
region. The first and second U-rich region and the A-rich region may associate
to form a triple helix on
the 3' end of the nucleic acid. This triple helix may stabilize the nucleic
acid, enhance the translational
efficiency of the nucleic acid and/or protect the 3' end from degradation.
Exemplary triple helices include,
but are not limited to, the triple helix sequence of metastasis-associated
lung adenocarcinoma transcript 1
(MALAT1), MEN-I3 and polyadenylated nuclear (PAN) RNA (See Wilusz et al.,
Genes & Development
2012 26:2392-2407). In one embodiment, the 3' end of the modified nucleic
acids, enhanced modified
RNA or ribonucleic acids of the present invention comprises a first U-rich
region comprising
TTTTTCTTTT (SEQ ID NO: 1), a second U-rich region comprising TTTTGCTTTTT (SEQ
ID NO: 2) or
TTTTGCTTTT (SEQ ID NO: 3), an A-rich region comprising AAAAAGCAAAA (SEQ ID NO:
4). In another
- 146 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
embodiment, the 3' end of the nucleic acids of the present invention comprises
a triple helix formation
structure comprising a first U-rich region, a conserved region, a second U-
rich region and an A-rich
region.
In one embodiment, the triple helix may be formed from the cleavage of a
MALAT1 sequence
prior to the cloverleaf structure. While not meaning to be bound by theory,
MALAT1 is a long non-coding
RNA which, when cleaved, forms a triple helix and a tRNA-like cloverleaf
structure. The MALAT1
transcript then localizes to nuclear speckles and the tRNA-like cloverleaf
localizes to the cytoplasm
(Wilusz et al. Cell 2008 135(5): 919-932).
As a non-limiting example, the terminal end of the nucleic acid of the present
invention
comprising the MALAT1 sequence can then form a triple helix structure, after
RNaseP cleavage from the
cloverleaf structure, which stabilizes the nucleic acid (Peart et al. Non-mRNA
3' end formation: how the
other half lives; WIREs RNA 2013).
In one embodiment, the nucleic acids or mRNA described herein comprise a
MALAT1
sequence. In another embodiment, the nucleic acids or mRNA may be
polyadenylated. In yet another
embodiment, the nucleic acids or mRNA is not polyadenylated but has an
increased resistance to
degradation compared to unmodified nucleic acids or mRNA.
In one embodiment, the nucleic acids of the present invention may comprise a
MALAT1
sequence in the second flanking region (e.g., the 3'UTR). As a non-limiting
example, the MALAT1
sequence may be human or mouse.
In another embodiment, the cloverleaf structure of the MALAT1 sequence may
also undergo
processing by RNaseZ and CCA adding enzyme to form a tRNA-like structure
called mascRNA
(MALAT1-associated small cytoplasmic RNA). As a non-limiting example, the
mascRNA may encode a
protein or a fragment thereof and/or may comprise a microRNA sequence. The
mascRNA may comprise
at least one chemical modification described herein.
Terminal Architecture Modifications: Poly-A tails
During RNA processing, a long chain of adenine nucleotides (poly-A tail) is
normally added to
a messenger RNA (mRNA) molecules to increase the stability of the molecule.
Immediately after
transcription, the 3 end of the transcript is cleaved to free a 3' hydroxyl.
Then poly-A polymerase adds a
chain of adenine nucleotides to the RNA. The process, called polyadenylation,
adds a poly-A tail that is
between 100 and 250 residues long.
Methods for the stabilization of RNA by incorporation of chain-terminating
nucleosides at the
3'-terminus include those described in International Patent Publication No.
W02013103659, the chain-
terminating nucleosides of which are incorporated herein.
Unique poly-A tail lengths may provide certain advantages to the modified RNAs
of the
present invention.
Generally, the length of a poly-A tail of the present invention is greater
than 30 nucleotides in
length. In another embodiment, the poly-A tail is greater than 35 nucleotides
in length. In another
embodiment, the length is at least 40 nucleotides. In another embodiment, the
length is at least 45
nucleotides. In another embodiment, the length is at least 55 nucleotides. In
another embodiment, the
length is at least 60 nucleotides. In another embodiment, the length is at
least 60 nucleotides. In another
- 147 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
embodiment, the length is at least 80 nucleotides. In another embodiment, the
length is at least 90
nucleotides. In another embodiment, the length is at least 100 nucleotides. In
another embodiment, the
length is at least 120 nucleotides. In another embodiment, the length is at
least 140 nucleotides. In
another embodiment, the length is at least 160 nucleotides. In another
embodiment, the length is at least
180 nucleotides. In another embodiment, the length is at least 200
nucleotides. In another embodiment,
the length is at least 250 nucleotides. In another embodiment, the length is
at least 300 nucleotides. In
another embodiment, the length is at least 350 nucleotides. In another
embodiment, the length is at least
400 nucleotides. In another embodiment, the length is at least 450
nucleotides. In another embodiment,
the length is at least 500 nucleotides. In another embodiment, the length is
at least 600 nucleotides. In
another embodiment, the length is at least 700 nucleotides. In another
embodiment, the length is at least
800 nucleotides. In another embodiment, the length is at least 900
nucleotides. In another embodiment,
the length is at least 1000 nucleotides. In another embodiment, the length is
at least 1100 nucleotides. In
another embodiment, the length is at least 1200 nucleotides. In another
embodiment, the length is at least
1300 nucleotides. In another embodiment, the length is at least 1400
nucleotides. In another
embodiment, the length is at least 1500 nucleotides. In another embodiment,
the length is at least 1600
nucleotides. In another embodiment, the length is at least 1700 nucleotides.
In another embodiment, the
length is at least 1800 nucleotides. In another embodiment, the length is at
least 1900 nucleotides. In
another embodiment, the length is at least 2000 nucleotides. In another
embodiment, the length is at least
2500 nucleotides. In another embodiment, the length is at least 3000
nucleotides.
In some embodiments, the nucleic acid or mRNA includes from about 30 to about
3,000
nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to
500, from 30 to 750, from 30
to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to
100, from 50 to 250, from 50 to
500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000,
from 50 to 2,500, from 50 to
3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500,
from 100 to 2,000, from
100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500
to 1,500, from 500 to
2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000
to 2,000, from 1,000 to
2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from
1,500 to 3,000, from 2,000 to
3,000, from 2,000 to 2,500, and from 2,500 to 3,000).
In one embodiment, the poly-A tail may be 80 nucleotides, 120 nucleotides, 160
nucleotides in
length on a modified RNA molecule described herein.
In another embodiment, the poly-A tail may be 20, 40, 80, 100, 120, 140 or 160
nucleotides in
length on a modified RNA molecule described herein.
In one embodiment, the poly-A tail is designed relative to the length of the
overall modified
RNA molecule. This design may be based on the length of the coding region of
the modified RNA, the
length of a particular feature or region of the modified RNA (such as the
mRNA), or based on the length
of the ultimate product expressed from the modified RNA. When relative to any
additional feature of the
modified RNA (e.g., other than the mRNA portion which includes the poly-A
tail) the poly-A tail may be 10,
20, 30, 40, 50, 60, 70, 80, 90 or 100% greater in length than the additional
feature. The poly-A tail may
also be designed as a fraction of the modified RNA to which it belongs. In
this context, the poly-A tail may
be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the
construct or the total length of
the construct minus the poly-A tail.
- 148 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In one embodiment, engineered binding sites and/or the conjugation of nucleic
acids or mRNA
for Poly-A binding protein may be used to enhance expression. The engineered
binding sites may be
sensor sequences which can operate as binding sites for ligands of the local
microenvironment of the
nucleic acids and/or mRNA. As a non-limiting example, the nucleic acids and/or
mRNA may comprise at
least one engineered binding site to alter the binding affinity of Poly-A
binding protein (PABP) and
analogs thereof. The incorporation of at least one engineered binding site may
increase the binding
affinity of the PABP and analogs thereof.
Additionally, multiple distinct nucleic acids or mRNA may be linked together
to the PABP
(Poly-A binding protein) through the 3'-end using modified nucleotides at the
3'-terminus of the poly-A tail.
Transfection experiments can be conducted in relevant cell lines at and
protein production can be
assayed by ELISA at 12hr, 24hr, 48hr, 72 hr and day 7 post-transfection. As a
non-limiting example, the
transfection experiments may be used to evaluate the effect on PABP or analogs
thereof binding affinity
as a result of the addition of at least one engineered binding site.
In one embodiment, a polyA tail may be used to modulate translation
initiation. While not
wishing to be bound by theory, the polyA til recruits PABP which in turn can
interact with translation
initiation complex and thus may be essential for protein synthesis.
In another embodiment, a polyA tail may also be used in the present invention
to protect
against 3'-5' exonuclease digestion.
In one embodiment, the nucleic acids or mRNA of the present invention are
designed to
include a polyA-G Quartet. The G-quartet is a cyclic hydrogen bonded array of
four guanine nucleotides
that can be formed by G-rich sequences in both DNA and RNA. In this
embodiment, the G-quartet is
incorporated at the end of the poly-A tail. The resultant nucleic acid or mRNA
may be assayed for
stability, protein production and other parameters including half-life at
various time points. It has been
discovered that the polyA-G quartet results in protein production equivalent
to at least 75% of that seen
using a poly-A tail of 120 nucleotides alone.
In one embodiment, the nucleic acids or mRNA of the present invention may
comprise a polyA
tail and may be stabilized by the addition of a chain terminating nucleoside.
The nucleic acids and/or
mRNA with a polyA tail may further comprise a 5'cap structure.
In another embodiment, the nucleic acids or mRNA of the present invention may
comprise a
polyA-G Quartet. The nucleic acids and/or mRNA with a polyA-G Quartet may
further comprise a 5'cap
structure.
In one embodiment, the chain terminating nucleoside which may be used to
stabilize the
nucleic acid or mRNA comprising a polyA tail or polyA-G Quartet may be, but is
not limited to, those
described in International Patent Publication No. W02013103659, the chain
terminating nucleosides of
which are incorporated herein by reference. In another embodiment, the chain
terminating nucleosides
which may be used with the present invention includes, but is not limited to,
3'-deoxyadenosine
(cordycepin), 3'-deoxyuridine, 3'-deoxycytosine, 3'-deoxyguanosine, 3'-
deoxythymine, 2',3'-
dideoxynucleosides, such as 2',3'- dideoxyadenosine, 2',3'-dideoxyuridine,
2',3'-dideoxycytosine, 2',3'-
dideoxyguanosine, 2',3'-dideoxythymine, a 2'-deoxynucleoside, or a -0-
methylnucleoside.
In another embodiment, the nucleic acid such as, but not limited to mRNA,
which comprise a
polyA tail or a polyA-G Quartet may be stabilized by a modification to the
3'region of the nucleic acid that
- 149 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
can prevent and/or inhibit the addition of oligio(U) (see e.g., International
Patent Publication No.
W02013103659, the modifications of which are incorporated herein by
reference).
In yet another embodiment, the nucleic acid such as, but not limited to mRNA,
which comprise
a polyA tail or a polyA-G Quartet may be stabilized by the addition of an
oligonucleotide that terminates in
a 3'-deoxynucleoside, 2',3'-dideoxynucleoside 3'-0- methylnucleosides, 3'-0-
ethylnucleosides, 3'-
arabinosides, and other modified nucleosides known in the art and/or described
herein.
5'UTR, 3'UTR and Translation Enhancer Elements (TEEs)
In one embodiment, the 5'UTR of the polynucleotides, primary constructs,
modified nucleic
acids and/or mmRNA may include at least one translational enhancer
polynucleotide, translation
enhancer element, translational enhancer elements (collectively referred to as
"TEE"s). As a non-limiting
example, the TEE may be located between the transcription promoter and the
start codon. The
polynucleotides, primary constructs, modified nucleic acids and/or mmRNA with
at least one TEE in the
5'UTR may include a cap at the 5'UTR. Further, at least one TEE may be located
in the 5'UTR of
polynucleotides, primary constructs, modified nucleic acids and/or mmRNA
undergoing cap-dependent or
cap-independent translation.
The term "translational enhancer element" or "translation enhancer element"
(herein
collectively referred to as "TEE") refers to sequences that increase the
amount of polypeptide or protein
produced from an mRNA.
In one aspect, TEEs are conserved elements in the UTR which can promote
translational
activity of a nucleic acid such as, but not limited to, cap-dependent or cap-
independent translation. The
conservation of these sequences has been previously shown by Panek et al
(Nucleic Acids Research,
2013, 1-10; incorporated herein by reference in its entirety) across 14
species including humans.
In one non-limiting example, the TEEs known may be in the 5'-leader of the Gtx
homeodomain
protein (Chappell et al., Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004).
In another non-limiting example, TEEs are disclosed as SEQ ID NOs: 1-35 in US
Patent
Publication No. U520090226470, SEQ ID NOs: 1-35 in US Patent Publication
U520130177581, SEQ ID
NOs: 1-35 in International Patent Publication No. W02009075886, SEQ ID NOs: 1-
5, and 7-645 in
International Patent Publication No. W02012009644, SEQ ID NO: 1 in
International Patent Publication
No. W01999024595, SEQ ID NO: 1 in US Patent No. U56310197, and SEQ ID NO: 1 in
US Patent No.
US6849405.
In yet another non-limiting example, the TEE may be an internal ribosome entry
site (IRES),
HCV-IRES or an IRES element such as, but not limited to, those described in US
Patent No. U57468275,
US Patent Publication Nos. U520070048776 and U520110124100 and International
Patent Publication
Nos. W02007025008 and W02001055369, the TEE sequences of each of which are
incorporated herein
by reference. The IRES elements may include, but are not limited to, the Gtx
sequences (e.g., Gtx9-nt,
Gtx8-nt, Gtx7-nt) described by Chappell et al. (Proc. Natl. Acad. Sci. USA
101:9590-9594, 2004) and
Zhou et al. (PNAS 102:6273-6278, 2005) and in US Patent Publication Nos.
U520070048776 and
U5201 10124100 and International Patent Publication No. W02007025008, the IRES
sequences of each
of which are incorporated herein by reference.
- 150 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
"Translational enhancer polynucleotides" or "translation enhancer
polynucleotide sequences"
are polynucleotides which include one or more of the specific TEE exemplified
herein and/or disclosed in
the art (see e.g., US6310197, US6849405, US7456273, US7183395, US20090226470,
US20070048776,
US20110124100, US20090093049, US20130177581, W02009075886, W02007025008,
W02012009644, W02001055371 W01999024595, and EP2610341A1 and EP2610340A1; the
TEE
sequences of each of which are incorporated herein by reference in its
entirety) or their variants,
homologs or functional derivatives. One or multiple copies of a specific TEE
can be present in the
polynucleotides, primary constructs, modified nucleic acids and/or mmRNA. The
TEEs in the translational
enhancer polynucleotides can be organized in one or more sequence segments. A
sequence segment
can harbor one or more of the specific TEEs exemplified herein, with each TEE
being present in one or
more copies. When multiple sequence segments are present in a translational
enhancer polynucleotide,
they can be homogenous or heterogeneous. Thus, the multiple sequence segments
in a translational
enhancer polynucleotide can harbor identical or different types of the
specific TEEs exemplified herein,
identical or different number of copies of each of the specific TEEs, and/or
identical or different
organization of the TEEs within each sequence segment.
In one embodiment, the polynucleotides, primary constructs, modified nucleic
acids and/or
mmRNA may include at least one TEE that is described in International Patent
Publication No.
W01999024595, W02012009644, W02009075886, W02007025008, W01999024595, European
Patent
Publication No. EP2610341A1 and EP2610340A1, US Patent No. US6310197,
US6849405, US7456273,
US7183395, US Patent Publication No. US20090226470, US20110124100,
US20070048776,
US20090093049, and US20130177581 the TEE sequences of each of which are
incorporated herein by
reference in its entirety. The TEE may be located in the 5'UTR of the
polynucleotides, primary constructs,
modified nucleic acids and/or mmRNA.
In another embodiment, the polynucleotides, primary constructs, modified
nucleic acids and/or
mmRNA may include at least one TEE that has at least 50%, at least 55%, at
least 60%, at least 65%, at
least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least
95% or at least 99% identity
with the TEEs described in US Patent Publication Nos. U520090226470,
U520070048776,
U520130177581 and U520110124100, International Patent Publication No.
W01999024595,
W02012009644, W02009075886 and W02007025008, European Patent Publication No.
EP2610341A1
and EP2610340A1, US Patent No. US6310197, U56849405, U57456273, US7183395, the
TEE
sequences of each of which are incorporated herein by reference in its
entirety.
In one embodiment, the 5'UTR of the polynucleotides, primary constructs,
modified nucleic
acids and/or mmRNA may include at least 1, at least 2, at least 3, at least 4,
at least 5, at least 6, at least
7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13,
at least 14, at least 15, at least 16,
at least 17, at least 18 at least 19, at least 20, at least 21, at least 22,
at least 23, at least 24, at least 25,
at least 30, at least 35, at least 40, at least 45, at least 50, at least 55
or more than 60 TEE sequences.
The TEE sequences in the 5'UTR of the polynucleotides, primary constructs,
modified nucleic acids
and/or mmRNA of the present invention may be the same or different TEE
sequences. The TEE
sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or
variants
thereof repeated once, twice, or more than three times. In these patterns,
each letter, A, B, or C
represent a different TEE sequence at the nucleotide level.
-151 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In one embodiment, the 5'UTR may include a spacer to separate two TEE
sequences. As a
non-limiting example, the spacer may be a 15 nucleotide spacer and/or other
spacers known in the art.
As another non-limiting example, the 5'UTR may include a TEE sequence-spacer
module repeated at
least once, at least twice, at least 3 times, at least 4 times, at least 5
times, at least 6 times, at least 7
times, at least 8 times and at least 9 times or more than 9 times in the
5'UTR.
In another embodiment, the spacer separating two TEE sequences may include
other
sequences known in the art which may regulate the translation of the
polynucleotides, primary constructs,
modified nucleic acids and/or mmRNA of the present invention such as, but not
limited to, miR sequences
described herein (e.g., miR binding sites and miR seeds). As a non-limiting
example, each spacer used to
separate two TEE sequences may include a different miR sequence or component
of a miR sequence
(e.g., miR seed sequence).
In one embodiment, the TEE in the 5'UTR of the polynucleotides, primary
constructs, modified
nucleic acids and/or mmRNA of the present invention may include at least 5%,
at least 10%, at least
15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at
least 45%, at least 50%, at
least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least
80%, at least 85%, at least
90%, at least 95%, at least 99% or more than 99% of the TEE sequences
disclosed in US Patent
Publication Nos. US20090226470, US20070048776, US20130177581 and
US20110124100,
International Patent Publication No. W01999024595, W02012009644, W02009075886
and
W02007025008, European Patent Publication No. EP2610341A1 and EP2610340A1, US
Patent No.
US6310197, US6849405, US7456273, and US7183395 the TEE sequences of each of
which are
incorporated herein by reference in its entirety. In another embodiment, the
TEE in the 5'UTR of the
polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of
the present invention may
include a 5-30 nucleotide fragment, a 5-25 nucleotide fragment, a 5-20
nucleotide fragment, a 5-15
nucleotide fragment, a 5-10 nucleotide fragment of the TEE sequences disclosed
in US Patent
Publication Nos. U520090226470, U520070048776, U520130177581 and
U520110124100,
International Patent Publication No. W01999024595, W02012009644, W02009075886
and
W02007025008, European Patent Publication No. EP2610341A1 and EP2610340A1, US
Patent No.
U56310197, U56849405, U57456273, and U57183395; each of which are incorporated
herein by
reference in its entirety.
In one embodiment, the TEE in the 5'UTR of the polynucleotides, primary
constructs, modified
nucleic acids and/or mmRNA of the present invention may include at least 5%,
at least 10%, at least
15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at
least 45%, at least 50%, at
least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least
80%, at least 85%, at least
90%, at least 95%, at least 99% or more than 99% of the TEE sequences
disclosed in Chappell et al.
(Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004) and Zhou et al. (PNAS
102:6273-6278, 2005), in
Supplemental Table 1 and in Supplemental Table 2 disclosed by Wellensiek et al
(Genome-wide profiling
of human cap-independent translation-enhancing elements, Nature Methods, 2013;
D01:10.1038/NMETH.2522). In another embodiment, the TEE in the 5'UTR of the
polynucleotides,
primary constructs, modified nucleic acids and/or mmRNA of the present
invention may include a 5-30
nucleotide fragment, a 5-25 nucleotide fragment, a 5-20 nucleotide fragment, a
5-15 nucleotide fragment,
a 5-10 nucleotide fragment of the TEE sequences disclosed in Chappell et al.
(Proc. Natl. Acad. Sci. USA
- 152 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
101:9590-9594, 2004) and Zhou et al. (PNAS 102:6273-6278, 2005), in
Supplemental Table 1 and in
Supplemental Table 2 disclosed by Wellensiek et al (Genome-wide profiling of
human cap-independent
translation-enhancing elements, Nature Methods, 2013; D01:10.1038/NMETH.2522);
the TEE sequences
of each of which are incorporated herein by reference in its entirety.
In one embodiment, the TEE used in the 5'UTR of the polynucleotides, primary
constructs,
modified nucleic acids and/or mmRNA of the present invention is an IRES
sequence such as, but not
limited to, those described in US Patent No. U57468275 and International
Patent Publication No.
W02001055369, the TEE sequences of each of which are incorporated herein by
reference in its entirety.
In one embodiment, the TEEs used in the 5'UTR of the polynucleotides, primary
constructs,
modified nucleic acids and/or mmRNA of the present invention may be identified
by the methods
described in US Patent Publication No. U520070048776 and U520110124100 and
International Patent
Publication Nos. W02007025008 and W02012009644, the TEE sequences of each of
which are
incorporated herein by reference in its entirety.
In another embodiment, the TEEs used in the 5'UTR of the polynucleotides,
primary
constructs, modified nucleic acids and/or mmRNA of the present invention may
be a transcription
regulatory element described in US Patent No. US7456273 and US7183395, US
Patent Publication No.
U520090093049, and International Publication No. W02001055371, the TEE
sequences of each of
which are incorporated herein by reference in its entirety. The transcription
regulatory elements may be
identified by methods known in the art, such as, but not limited to, the
methods described in US Patent
No. U57456273 and US7183395, US Patent Publication No. U520090093049, and
International
Publication No. W02001055371, the transcription regulatory elements of each of
which are incorporated
herein by reference in its entirety.
In yet another embodiment, the TEE used in the 5'UTR of the polynucleotides,
primary
constructs, modified nucleic acids and/or mmRNA of the present invention is an
oligonucleotide or portion
thereof as described in US Patent No. U57456273 and US7183395, US Patent
Publication No.
U520090093049, and International Publication No. W02001055371, the TEE
sequences of each of
which are incorporated herein by reference in its entirety.
The 5' UTR comprising at least one TEE described herein may be incorporated in
a
monocistronic sequence such as, but not limited to, a vector system or a
nucleic acid vector. As a non-
limiting example, the vector systems and nucleic acid vectors may include
those described in US Patent
Nos. 7456273 and US7183395, US Patent Publication No. U520070048776,
U520090093049 and
U520110124100 and International Patent Publication Nos. W02007025008 and
W02001055371, the
TEE sequences of each of which are incorporated herein by reference in its
entirety.
In one embodiment, the TEEs described herein may be located in the 5'UTR
and/or the 3'UTR
of the polynucleotides, primary constructs, modified nucleic acids and/or
mmRNA. The TEEs located in
the 3'UTR may be the same and/or different than the TEEs located in and/or
described for incorporation
in the 5'UTR.
In one embodiment, the 3'UTR of the polynucleotides, primary constructs,
modified nucleic
acids and/or mmRNA may include at least 1, at least 2, at least 3, at least 4,
at least 5, at least 6, at least
7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13,
at least 14, at least 15, at least 16,
at least 17, at least 18 at least 19, at least 20, at least 21, at least 22,
at least 23, at least 24, at least 25,
- 153 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
at least 30, at least 35, at least 40, at least 45, at least 50, at least 55
or more than 60 TEE sequences.
The TEE sequences in the 3'UTR of the polynucleotides, primary constructs,
modified nucleic acids
and/or mmRNA of the present invention may be the same or different TEE
sequences. The TEE
sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or
variants
thereof repeated once, twice, or more than three times. In these patterns,
each letter, A, B, or C
represent a different TEE sequence at the nucleotide level.
In one embodiment, the 3'UTR may include a spacer to separate two TEE
sequences. As a
non-limiting example, the spacer may be a 15 nucleotide spacer and/or other
spacers known in the art.
As another non-limiting example, the 3'UTR may include a TEE sequence-spacer
module repeated at
least once, at least twice, at least 3 times, at least 4 times, at least 5
times, at least 6 times, at least 7
times, at least 8 times and at least 9 times or more than 9 times in the
3'UTR.
In another embodiment, the spacer separating two TEE sequences may include
other
sequences known in the art which may regulate the translation of the
polynucleotides, primary constructs,
modified nucleic acids and/or mmRNA of the present invention such as, but not
limited to, miR sequences
described herein (e.g., miR binding sites and miR seeds). As a non-limiting
example, each spacer used
to separate two TEE sequences may include a different miR sequence or
component of a miR sequence
(e.g., miR seed sequence).
In one embodiment, the incorporation of a miR sequence and/or a TEE sequence
changes the
shape of the stem loop region which may increase and/or decrease translation.
(see e.g, Kedde et al. A
Pumilio-induced RNA structure switch in p27-3'UTR controls miR-221 and miR-22
accessibility. Nature
Cell Biology. 2010).
Heterologous 5'UTRs
A 5' UTR may be provided as a flanking region to the modified nucleic acids
(mRNA),
enhanced modified RNA or ribonucleic acids of the invention. 5'UTR may be
homologous or heterologous
to the coding region found in the modified nucleic acids (mRNA), enhanced
modified RNA or ribonucleic
acids of the invention. Multiple 5' UTRs may be included in the flanking
region and may be the same or of
different sequences. Any portion of the flanking regions, including none, may
be codon optimized and
any may independently contain one or more different structural or chemical
modifications, before and/or
after codon optimization.
Shown in Lengthy Table 21 in US Provisional Application No 61/775,509, and in
Lengthy
Table 21 and in Table 22 in US Provisional Application No 61/829,372, the
sequences of which are
incorporated herein by reference in their entirety, is a listing of the start
and stop site of the modified
nucleic acids (mRNA), enhanced modified RNA or ribonucleic acids of the
invention. In Table 21 each
5'UTR (5'UTR-005 to 5'UTR 68511) is identified by its start and stop site
relative to its native or wild type
(homologous) transcript (ENST; the identifier used in the ENSEMBL database).
To alter one or more properties of the polynucleotides, primary constructs or
mmRNA of the
invention, 5'UTRs which are heterologous to the coding region of the modified
nucleic acids (mRNA),
enhanced modified RNA or ribonucleic acids of the invention are engineered
into compounds of the
invention. The modified nucleic acids (mRNA), enhanced modified RNA or
ribonucleic acids are then
administered to cells, tissue or organisms and outcomes such as protein level,
localization and/or half-life
- 154 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
are measured to evaluate the beneficial effects the heterologous 5'UTR may
have on the modified nucleic
acids (mRNA), enhanced modified RNA or ribonucleic acids of the invention.
Variants of the 5' UTRs
may be utilized wherein one or more nucleotides are added or removed to the
termini, including A, T, C or
G. 5'UTRs may also be codon-optimized or modified in any manner described
herein.
Incorporating microRNA Binding Sites
In one embodiment, modified nucleic acids (mRNA), enhanced modified RNA or
ribonucleic
acids of the invention would not only encode a polypeptide but also a sensor
sequence. Sensor
sequences include, for example, microRNA binding sites, transcription factor
binding sites, structured
mRNA sequences and/or motifs, artificial binding sites engineered to act as
pseudo-receptors for
endogenous nucleic acid binding molecules. Non-limiting examples, of
polynucleotides comprising at
least one sensor sequence are described in co-pending and co-owned U.S.
Provisional Patent
Application No. US 61/753,661, filed January 17, 2013, U.S. Provisional Patent
Application No. US
61/754,159, filed January 18, 2013, U.S. Provisional Patent Application No.
U561/781 097, filed March
14, 2013, U.S. Provisional Patent Application No. US 61/829,334, filed May 31,
2013, U.S. Provisional
Patent Application No. US 61/839,893, filed June 27, 2013, U.S. Provisional
Patent Application No. US
61/842,733, filed July 3, 2013, and US Provisional Patent Application No. US
61/857,304, filed July 23,
2013, the sequences of each of which are incorporated herein by reference in
their entirety.
In one embodiment, microRNA (miRNA) profiling of the target cells or tissues
is conducted to
determine the presence or absence of miRNA in the cells or tissues.
microRNAs (or miRNA) are 19-25 nucleotide long noncoding RNAs that bind to the
3'UTR of
nucleic acid molecules and down-regulate gene expression either by reducing
nucleic acid molecule
stability or by inhibiting translation. The modified nucleic acids (mRNA),
enhanced modified RNA or
ribonucleic acids of the invention may comprise one or more microRNA target
sequences, microRNA
sequences, or microRNA seeds. Such sequences may correspond to any known
microRNA such as
those taught in US Publication U52005/0261218 and US Publication
U52005/0059005, the miRNAs of
which are incorporated herein by reference in their entirety.
A microRNA sequence comprises a "seed" region, i.e., a sequence in the region
of positions
2-8 of the mature microRNA, which sequence has perfect Watson-Crick
complementarity to the miRNA
target sequence. A microRNA seed may comprise positions 2-8 or 2-7 of the
mature microRNA. In some
embodiments, a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8
of the mature
microRNA), wherein the seed-complementary site in the corresponding miRNA
target is flanked by an
adenine (A) opposed to microRNA position 1. In some embodiments, a microRNA
seed may comprise 6
nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-
complementary site in the
corresponding miRNA target is flanked by an adenine (A) opposed to microRNA
position 1. See for
example, Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP;
Mol Cell. 2007 Jul
6;27(1):91-105. The bases of the microRNA seed have complete complementarity
with the target
sequence. By engineering microRNA target sequences into the 3'UTR of nucleic
acids or mRNA of the
invention one can target the molecule for degradation or reduced translation,
provided the microRNA in
question is available. This process will reduce the hazard of off target
effects upon nucleic acid molecule
delivery. Identification of microRNA, microRNA target regions, and their
expression patterns and role in
- 155 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
biology have been reported (Bonauer et al., Curr Drug Targets 2010 11:943-949;
Anand and Cheresh
Curr Opin Hematol 2011 18:171-176; Contreras and Rao Leukemia 2012 26:404-413
(2011 Dec 20. doi:
10.1038/Ieu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell,
2007 129:1401-1414; Gentner
and Naldini, Tissue Antigens. 2012 80:393-403 and all references therein; the
miRNAs of each of which is
incorporated herein by reference in its entirety).
For example, if the mRNA is not intended to be delivered to the liver but ends
up there, then
miR-122, a microRNA abundant in liver, can inhibit the expression of the gene
of interest if one or multiple
target sites of miR-122 are engineered into the 3'UTR of the modified nucleic
acids, enhanced modified
RNA or ribonucleic acids. Introduction of one or multiple binding sites for
different microRNA can be
engineered to further decrease the longevity, stability, and protein
translation of a modified nucleic acids,
enhanced modified RNA or ribonucleic acids. As used herein, the term "microRNA
site" refers to a
microRNA target site or a microRNA recognition site, or any nucleotide
sequence to which a microRNA
binds or associates. It should be understood that "binding" may follow
traditional Watson-Crick
hybridization rules or may reflect any stable association of the microRNA with
the target sequence at or
adjacent to the microRNA site.
Conversely, for the purposes of the modified nucleic acids, enhanced modified
RNA or
ribonucleic acids of the present invention, microRNA binding sites can be
engineered out of (i.e. removed
from) sequences in which they naturally occur in order to increase protein
expression in specific tissues.
For example, miR-122 binding sites may be removed to improve protein
expression in the liver.
In one embodiment, the modified nucleic acids, enhanced modified RNA or
ribonucleic acids
of the present invention may include at least one miRNA-binding site in the
3'UTR in order to direct
cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but
not limited to, normal and/or
cancerous cells (e.g., HEP3B or SNU449).
In another embodiment, the modified nucleic acids, enhanced modified RNA or
ribonucleic
acids of the present invention may include three miRNA-binding sites in the
3'UTR in order to direct
cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but
not limited to, normal and/or
cancerous cells (e.g., HEP3B or SNU449).
Regulation of expression in multiple tissues can be accomplished through
introduction or
removal or one or several microRNA binding sites. The decision of removal or
insertion of microRNA
binding sites, or any combination, is dependent on microRNA expression
patterns and their profilings in
diseases.
Examples of tissues where microRNA are known to regulate mRNA, and thereby
protein
expression, include, but are not limited to, liver (miR-122), muscle (miR-133,
miR-206, miR-208),
endothelial cells (miR-17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p,
miR-16, miR-21, miR-
223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-1d, miR-
149), kidney (miR-192, miR-
194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126).
Specifically, microRNAs are known to be differentially expressed in immune
cells (also called
hematopoietic cells), such as antigen presenting cells (APCs) (e.g. dendritic
cells and macrophages),
macrophages, monocytes, B lymphocytes, T lymphocytes, granuocytes, natural
killer cells, etc. Immune
cell specific microRNAs are involved in immunogenicity, autoimmunity, the
immune -response to
infection, inflammation, as well as unwanted immune response after gene
therapy and tissue/organ
- 156 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
transplantation. Immune cells specific microRNAs also regulate many aspects of
development,
proliferation, differentiation and apoptosis of hematopoietic cells (immune
cells). For example, miR-142
and miR-146 are exclusively expressed in the immune cells, particularly
abundant in myeloid dendritic
cells. It was demonstrated in the art that the immune response to exogenous
nucleic acid molecules was
shut-off by adding miR-142 binding sites to the 3'UTR of the delivered gene
construct, enabling more
stable gene transfer in tissues and cells. miR-142 efficiently degrades the
exogenous mRNA in antigen
presenting cells and suppresses cytotoxic elimination of transduced cells
(Annoni A et al., blood, 2009,
114, 5152-5161; Brown BD, et al., Nat med. 2006, 12(5), 585-591; Brown BD, et
al., blood, 2007,
110(13): 4144-4152, the miRNAs of each of which is incorporated herein by
reference in its entirety).
An antigen-mediated immune response can refer to an immune response triggered
by foreign
antigens, which, when entering an organism, are processed by the antigen
presenting cells and displayed
on the surface of the antigen presenting cells. T cells can recognize the
presented antigen and induce a
cytotoxic elimination of cells that express the antigen.
Introducing the miR-142 binding site into the 3'-UTR of a polypeptide of the
present invention
can selectively repress the gene expression in the antigen presenting cells
through miR-142 mediated
mRNA degradation, limiting antigen presentation in APCs (e.g. dendritic cells)
and thereby preventing
antigen-mediated immune response after the delivery of the polynucleotides.
The polynucleotides are
therefore stably expressed in target tissues or cells without triggering
cytotoxic elimination.
In one embodiment, microRNAs binding sites that are known to be expressed in
immune cells,
in particular, the antigen presenting cells, can be engineered into the
polynucleotide to suppress the
expression of the sensor-signal polynucleotide in APCs through microRNA
mediated RNA degradation,
subduing the antigen-mediated immune response, while the expression of the
polynucleotide is
maintained in non-immune cells where the immune cell specific microRNAs are
not expressed. For
example, to prevent the immunogenic reaction caused by a liver specific
protein expression, the miR-122
binding site can be removed and the miR-142 (and/or mirR-146) binding sites
can be engineered into the
3-UTR of the polynucleotide.
To further drive the selective degradation and suppression of mRNA in APCs and
macrophage, the polynucleotide may include another negative regulatory element
in the 3-UTR, either
alone or in combination with mir-142 and/or mir-146 binding sites. As a non-
limiting example, one
regulatory element is the Constitutive Decay Elements (CDEs).
Immune cells specific microRNAs include, but are not limited to, hsa-let-7a-2-
3p, hsa-let-7a-
3p, hsa-7a-5p, hsa-let-7c, hsa-let-7e-3p, hsa-let-7e-5p, hsa-let-7g-3p, hsa-
let-7g-5p, hsa-let-7i-3p, hsa-
let-7i-5p, miR-10a-3p, miR-10a-5p, miR-1184, hsa-let-7f-1--3p, hsa-let-7f-2--
5p, hsa-let-7f-5p, miR-125b-
1-3p, miR-125b-2-3p, miR-125b-5p, miR-1279, miR-130a-3p, miR-130a-5p, miR-132-
3p, miR-132-5p,
miR-142-3p, miR-142-5p, miR-143-3p, miR-143-5p, miR-146a-3p, miR-146a-5p, miR-
146b-3p, miR-
146b-5p, miR-147a, miR-147b, miR-148a-5p, miR-148a-3p, miR-150-3p, miR-150-5p,
miR-151b, miR-
155-3p, miR-155-5p, miR-15a-3p, miR-15a-5p, miR-15b-5p, miR-15b-3p, miR-16-1-
3p, miR-16-2-3p,
miR-16-5p, miR-17-5p, miR-181a-3p, miR-181a-5p, miR-181a-2-3p, miR-182-3p, miR-
182-5p, miR-197-
3p, miR-197-5p, miR-21-5p, miR-21-3p, miR-214-3p, miR-214-5p, miR-223-3p, miR-
223-5p, miR-221-
3p, miR-221-5p, miR-23b-3p, miR-23b-5p, miR-24-1-5p,miR-24-2-5p, miR-24-3p,
miR-26a-1-3p, miR-
26a-2-3p, miR-26a-5p, miR-26b-3p, miR-26b-5p, miR-27a-3p, miR-27a-5p, miR-27b-
3p,miR-27b-5p,
- 157 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
miR-28-3p, miR-28-5p, miR-2909, miR-29a-3p, miR-29a-5p, miR-29b-1-5p, miR-29b-
2-5p, miR-29c-3p,
miR-29c-5põ miR-30e-3p, miR-30e-5p, miR-331-5p, miR-339-3p, miR-339-5p, miR-
345-3p, miR-345-5p,
miR-346, miR-34a-3p, miR-34a-5põ miR-363-3p, miR-363-5p, miR-372, miR-377-3p,
miR-377-5p, miR-
493-3p, miR-493-5p, miR-542, miR-548b-5p, miR548c-5p, miR-548i, miR-548j, miR-
548n, miR-574-3p,
miR-598, miR-718, miR-935, miR-99a-3p, miR-99a-5p, miR-99b-3p and miR-99b-5p.
Furthermore, novel
miroRNAs are discovered in the immune cells in the art through micro-array
hybridization and microtome
analysis (Jima DD et al, Blood, 2010, 116:e118-e127; Vaz C et al., BMC
Genomics, 2010, 11,288, the
miRNAs of each of which is incorporated herein by reference in its entirety.)
MicroRNAs that are known to be expressed in the liver include, but are not
limited to, miR-107,
miR-122-3p, miR-122-5p, miR-1228-3p, miR-1228-5p, miR-1249, miR-129-5p, miR-
1303, miR-151a-3p,
miR-151a-5p, miR-152, miR-194-3p, miR-194-5p, miR-199a-3p, miR-199a-5p, miR-
199b-3p, miR-199b-
5p, miR-296-5p, miR-557, miR-581, miR-939-3p, miR-939-5p. MicroRNA binding
sites from any liver
specific microRNA can be introduced to or removed from the polynucleotides to
regulate the expression
of the polynucleotides in the liver. Liver specific microRNAs binding sites
can be engineered alone or
further in combination with immune cells (e.g. APCs) microRNA binding sites in
order to prevent immune
reaction against protein expression in the liver.
MicroRNAs that are known to be expressed in the lung include, but are not
limited to, let-7a-2-
3p, let-7a-3p, let-7a-5p, miR-126-3p, miR-126-5p, miR-127-3p, miR-127-5p, miR-
130a-3p, miR-130a-5p,
miR-130b-3p, miR-130b-5p, miR-133a, miR-133b, miR-134, miR-18a-3p, miR-18a-5p,
miR-18b-3p, miR-
18b-5p, miR-24-1-5p, miR-24-2-5p, miR-24-3p, miR-296-3p, miR-296-5p, miR-32-
3p, miR-337-3p, miR-
337-5p, miR-381-3p, miR-381-5p. MicroRNA binding sites from any lung specific
microRNA can be
introduced to or removed from the polynucleotide to regulate the expression of
the polynucleotide in the
lung. Lung specific microRNAs binding sites can be engineered alone or further
in combination with
immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune
reaction against protein
expression in the lung.
MicroRNAs that are known to be expressed in the heart include, but are not
limited to, miR-1,
miR-133a, miR-133b, miR-149-3p, miR-149-5p, miR-186-3p, miR-186-5p, miR-208a,
miR-208b, miR-210,
miR-296-3p, miR-320, miR-451a, miR-451b, miR-499a-3p, miR-499a-5p, miR-499b-
3p, miR-499b-5p,
miR-744-3p, miR-744-5p, miR-92b-3p and miR-92b-5p. MicroRNA binding sites from
any heart specific
microRNA can be introduced to or removed from the polynucleotides to regulate
the expression of the
polynucleotides in the heart. Heart specific microRNAs binding sites can be
engineered alone or further in
combination with immune cells (e.g. APCs) microRNA binding sites to prevent an
immune reaction
against protein expression in the heart.
MicroRNAs that are known to be expressed in the nervous system include, but
are not limited
to, miR-124-5p, miR-125a-3p, miR-125a-5p, miR-125b-1-3p, miR-125b-2-3p, miR-
125b-5p,miR-1271-3p,
miR-1271-5p, miR-128, miR-132-5p, miR-135a-3p, miR-135a-5p, miR-135b-3p, miR-
135b-5p, miR-137,
miR-139-5p, miR-139-3p, miR-149-3p, miR-149-5p, miR-153, miR-181c-3p, miR-181c-
5p, miR-183-3p,
miR-183-5p, miR-190a, miR-190b, miR-212-3p, miR-212-5p, miR-219-1-3p, miR-219-
2-3p, miR-23a-3p,
miR-23a-5p,miR-30a-5p, miR-30b-3p, miR-30b-5p, miR-30c-1-3p, miR-30c-2-3p, miR-
30c-5p, miR-30d-
3p, miR-30d-5p, miR-329, miR-342-3p, miR-3665, miR-3666, miR-380-3p, miR-380-
5p, miR-383, miR-
410, miR-425-3p, miR-425-5p, miR-454-3p, miR-454-5p, miR-483, miR-510, miR-
516a-3p, miR-548b-5p,
- 158 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
miR-548c-5p, miR-571, miR-7-1-3p, miR-7-2-3p, miR-7-5p, miR-802, miR-922, miR-
9-3p and miR-9-5p.
MicroRNAs enriched in the nervous system further include those specifically
expressed in neurons,
including, but not limited to, miR-132-3p, miR-132-3p, miR-148b-3p, miR-148b-
5p, miR-151a-3p, miR-
151a-5p, miR-212-3p, miR-212-5p, miR-320b, miR-320e, miR-323a-3p, miR-323a-5p,
miR-324-5p, miR-
325, miR-326, miR-328, miR-922 and those specifically expressed in glial
cells, including, but not limited
to, miR-1250, miR-219-1-3p, miR-219-2-3p, miR-219-5p, miR-23a-3p, miR-23a-5p,
miR-3065-3p, miR-
3065-5p, miR-30e-3p, miR-30e-5p, miR-32-5p, miR-338-5p, miR-657. MicroRNA
binding sites from any
CNS specific microRNA can be introduced to or removed from the polynucleotides
to regulate the
expression of the polynucleotide in the nervous system. Nervous system
specific microRNAs binding
sites can be engineered alone or further in combination with immune cells
(e.g. APCs) microRNA binding
sites in order to prevent immune reaction against protein expression in the
nervous system.
MicroRNAs that are known to be expressed in the pancreas include, but are not
limited to,
miR-105-3p, miR-105-5p, miR-184, miR-195-3p, miR-195-5p, miR-196a-3p, miR-196a-
5p, miR-214-3p,
miR-214-5p, miR-216a-3p, miR-216a-5p, miR-30a-3p, miR-33a-3p, miR-33a-5p, miR-
375, miR-7-1-3p,
miR-7-2-3p, miR-493-3p, miR-493-5p and miR-944. MicroRNA binding sites from
any pancreas specific
microRNA can be introduced to or removed from the polynucleotide to regulate
the expression of the
polynucleotide in the pancreas. Pancreas specific microRNAs binding sites can
be engineered alone or
further in combination with immune cells (e.g. APCs) microRNA binding sites in
order to prevent an
immune reaction against protein expression in the pancreas.
MicroRNAs that are known to be expressed in the kidney further include, but
are not limited to,
miR-122-3p, miR-145-5p, miR-17-5p, miR-192-3p, miR-192-5p, miR-194-3p, miR-194-
5p, miR-20a-3p,
miR-20a-5p, miR-204-3p, miR-204-5p, miR-210, miR-216a-3p, miR-216a-5p, miR-296-
3p, miR-30a-3p,
miR-30a-5p, miR-30b-3p, miR-30b-5p, miR-30c-1-3p, miR-30c-2-3p, miR30c-5p, miR-
324-3p, miR-335-
3p, miR-335-5p, miR-363-3p, miR-363-5p and miR-562. MicroRNA binding sites
from any kidney specific
microRNA can be introduced to or removed from the polynucleotide to regulate
the expression of the
polynucleotide in the kidney. Kidney specific microRNAs binding sites can be
engineered alone or further
in combination with immune cells (e.g. APCs) microRNA binding sites to prevent
an immune reaction
against protein expression in the kidney.
MicroRNAs that are known to be expressed in the muscle further include, but
are not limited
to, let-7g-3p, let-7g-5p, miR-1, miR-1286, miR-133a, miR-133b, miR-140-3p, miR-
143-3p, miR-143-5p,
miR-145-3p, miR-145-5p, miR-188-3p, miR-188-5p, miR-206, miR-208a, miR-208b,
miR-25-3p and miR-
25-5p. MicroRNA binding sites from any muscle specific microRNA can be
introduced to or removed from
the polynucleotide to regulate the expression of the polynucleotide in the
muscle. Muscle specific
microRNAs binding sites can be engineered alone or further in combination with
immune cells (e.g.
APCs) microRNA binding sites to prevent an immune reaction against protein
expression in the muscle.
MicroRNAs are differentially expressed in different types of cells, such as
endothelial cells,
epithelial cells and adipocytes. For example, microRNAs that are expressed in
endothelial cells include,
but are not limited to, let-7b-3p, let-7b-5p, miR-100-3p, miR-100-5p, miR-101-
3p, miR-101-5p, miR-126-
3p, miR-126-5p, miR-1236-3p, miR-1236-5p, miR-130a-3p, miR-130a-5p, miR-17-5p,
miR-17-3p, miR-
18a-3p, miR-18a-5põ miR-19a-3p, miR-19a-5p, miR-19b-1-5p, miR-19b-2-5p, miR-
19b-3p, miR-20a-3p,
miR-20a-5p, miR-217, miR-210, miR-21-3p, miR-21-5p, miR-221-3p, miR-221-5p,
miR-222-3p, miR-222-
- 159 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
5p, miR-23a-3p, miR-23a-5p, miR-296-5p, miR-361-3p, miR-361-5p, miR-421, miR-
424-3p, miR-424-5p,
miR-513a-5p, miR-92a-1-5p, miR-92a-2-5p, miR-92a-3p, miR-92b-3p and miR-92b-
5p. Many novel
microRNAs are discovered in endothelial cells from deep-sequencing analysis
(Voellenkle C et al., RNA,
2012, 18, 472-484, the miRNAs of which are herein incorporated by reference)
microRNA binding sites
from any endothelial cell specific microRNA can be introduced to or removed
from the polynucleotide to
modulate the expression of the polynucleotide in the endothelial cells in
various conditions.
For further example, microRNAs that are expressed in epithelial cells include,
but are not
limited to, let-7b-3p, let-7b-5p, miR-1246, miR-200a-3p, miR-200a-5p, miR-200b-
3p, miR-200b-5p, miR-
200c-3p, miR-200c-5p, miR-338-3p, miR-429, miR-451a, miR-451b, miR-494, miR-
802 and miR-34a,
miR-34b-5p , miR-34c-5p, miR-449a, miR-449b-3p, miR-449b-5p specific in
respiratory ciliated epithelial
cells; let-7 family, miR-133a, miR-133b, miR-126 specific in lung epithelial
cells; miR-382-3p, miR-382-5p
specific in renal epithelial cells and miR-762 specific in corneal epithelial
cells. MicroRNA binding sites
from any epithelial cell specific MicroRNA can be introduced to or removed
from the polynucleotide to
modulate the expression of the polynucleotide in the epithelial cells in
various conditions.
In addition, a large group of microRNAs are enriched in embryonic stem cells,
controlling stem
cell self-renewal as well as the development and/or differentiation of various
cell lineages, such as neural
cells, cardiac, hematopoietic cells, skin cells, osteogenic cells and muscle
cells (Kuppusamy KT et al.,
Curr. Mol Med, 2013, 13(5), 757-764; Vidigal JA and Ventura A, Semin Cancer
Biol. 2012, 22(5-6), 428-
436; Goff LA et al., PLoS One, 2009, 4:e7192; Morin RD et al., Genome
Res,2008,18, 610-621; Yoo JK
et al., Stem Cells Dev. 2012, 21(11), 2049-2057, the miRNAs of each of which
is herein incorporated by
reference in its entirety). MicroRNAs abundant in embryonic stem cells
include, but are not limited to, let-
7a-2-3p, let-a-3p, let-7a-5p, let7d-3p, let-7d-5p, miR-103a-2-3p, miR-103a-5p,
miR-106b-3p, miR-106b-
5p, miR-1246, miR-1275, miR-138-1-3p, miR-138-2-3p, miR-138-5p, miR-154-3p,
miR-154-5p, miR-200c-
3p, miR-200c-5p, miR-290, miR-301a-3p, miR-301a-5p, miR-302a-3p, miR-302a-5p,
miR-302b-3p, miR-
302b-5p, miR-302c-3p, miR-302c-5p, miR-302d-3p, miR-302d-5p, miR-302e, miR-367-
3p, miR-367-5p,
miR-369-3p, miR-369-5p, miR-370, miR-371, miR-373, miR-380-5p, miR-423-3p, miR-
423-5p, miR-486-
5p, miR-520c-3p, miR-548e, miR-548f, miR-548g-3p, miR-548g-5p, miR-548i, miR-
548k, miR-548I, miR-
548m, miR-548n, miR-548o-3p, miR-548o-5p, miR-548p, miR-664a-3p, miR-664a-5p,
miR-664b-3p, miR-
664b-5p, miR-766-3p, miR-766-5p, miR-885-3p, miR-885-5p,miR-93-3p, miR-93-5p,
miR-941,miR-96-
3p, miR-96-5p, miR-99b-3p and miR-99b-5p. Many predicted novel microRNAs are
discovered by deep
sequencing in human embryonic stem cells (Morin RD et al., Genome Res,2008,18,
610-621; Goff LA et
al., PLoS One, 2009, 4:e7192; Bar M et al., Stem cells, 2008, 26, 2496-2505,
the miRNAs of each of
which is incorporated herein by references).
In one embodiment, the binding sites of embryonic stem cell specific microRNAs
can be
included in or removed from the 3-UTR of the polynucleotide to modulate the
development and/or
differentiation of embryonic stem cells, to inhibit the senescence of stem
cells in a degenerative condition
(e.g. degenerative diseases), or to stimulate the senescence and apoptosis of
stem cells in a disease
condition (e.g. cancer stem cells).
Many microRNA expression studies are conducted in the art to profile the
differential
expression of microRNAs in various cancer cells /tissues and other diseases.
Some microRNAs are
abnormally over-expressed in certain cancer cells and others are under-
expressed. For example,
- 160 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
microRNAs are differentially expressed in cancer cells (W02008/154098,
US2013/0059015,
US2013/0042333, W02011/157294); cancer stem cells (US2012/0053224); pancreatic
cancers and
diseases (US2009/0131348, US2011/0171646, US2010/0286232, US8389210); asthma
and
inflammation (US8415096); prostate cancer (US2013/0053264); hepatocellular
carcinoma
(W02012/151212, US2012/0329672, W02008/054828, US8252538); lung cancer cells
(W02011/076143, W02013/033640, W02009/070653, US2010/0323357); cutaneous T
cell lymphoma
(W02013/011378); colorectal cancer cells (W02011/0281756, W02011/076142);
cancer positive lymph
nodes (W02009/100430, US2009/0263803); nasopharyngeal carcinoma (EP2112235);
chronic
obstructive pulmonary disease (US2012/0264626, US2013/0053263); thyroid cancer
(W02013/066678);
ovarian cancer cells ( US2012/0309645, W02011/095623); breast cancer cells
(W02008/154098,
W02007/081740, US2012/0214699), leukemia and lymphoma (W02008/073915,
US2009/0092974,
US2012/0316081, US2012/0283310, W02010/018563, the miRNAs of each of which is
incorporated
herein by reference.)
As a non-limiting example, microRNA sites that are over-expressed in certain
cancer and/or
tumor cells can be removed from the 3-UTR of the polynucleotide encoding the
polypeptide of interest,
restoring the expression suppressed by the over-expressed microRNAs in cancer
cells, thus ameliorating
the corresponsive biological function, for instance, transcription stimulation
and/or repression, cell cycle
arrest, apoptosis and cell death. Normal cells and tissues, wherein microRNAs
expression is not up-
regulated, will remain unaffected.
MicroRNA can also regulate complex biological processes such as angiogenesis
(miR-132)
(Anand and Cheresh Curr Opin Hematol 2011 18:171-176). In the modified nucleic
acids, enhanced
modified RNA or ribonucleic acids of the invention, binding sites for
microRNAs that are involved in such
processes may be removed or introduced, in order to tailor the expression of
the modified nucleic acids,
enhanced modified RNA or ribonucleic acids expression to biologically relevant
cell types or to the
context of relevant biological processes. In this context, the mRNA are
defined as auxotrophic mRNA.
MicroRNA gene regulation may be influenced by the sequence surrounding the
microRNA
such as, but not limited to, the species of the surrounding sequence, the type
of sequence (e.g.,
heterologous, homologous and artificial), regulatory elements in the
surrounding sequence and/or
structural elements in the surrounding sequence. The microRNA may be
influenced by the 5'UTR and/or
the 3'UTR. As a non-limiting example, a non-human 3'UTR may increase the
regulatory effect of the
microRNA sequence on the expression of a polypeptide of interest compared to a
human 3'UTR of the
same sequence type.
In one embodiment, other regulatory elements and/or structural elements of the
5'-UTR can
influence microRNA mediated gene regulation. One example of a regulatory
element and/or structural
element is a structured IRES (Internal Ribosome Entry Site) in the 5'UTR,
which is necessary for the
binding of translational elongation factors to initiate protein translation.
ElF4A2 binding to this secondarily
structured element in the 5'UTR is necessary for microRNA mediated gene
expression (Meijer HA et al.,
Science, 2013, 340, 82-85, herein incorporated by reference in its entirety).
The modified nucleic acids,
enhanced modified RNA or ribonucleic acids of the invention can further be
modified to include this
structured 5'-UTR in order to enhance microRNA mediated gene regulation.
- 161 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
At least one microRNA site can be engineered into the 3' UTR of the modified
nucleic acids,
enhanced modified RNA or ribonucleic acids of the present invention. In this
context, at least two, at least
three, at least four, at least five, at least six, at least seven, at least
eight, at least nine, at least ten or
more microRNA sites may be engineered into the 3' UTR of the ribonucleic acids
of the present invention.
In one embodiment, the microRNA sites incorporated into the modified nucleic
acids, enhanced modified
RNA or ribonucleic acids may be the same or may be different microRNA sites.
In another embodiment,
the microRNA sites incorporated into the modified nucleic acids, enhanced
modified RNA or ribonucleic
acids may target the same or different tissues in the body. As a non-limiting
example, through the
introduction of tissue-, cell-type-, or disease-specific microRNA binding
sites in the 3' UTR of a modified
nucleic acid mRNA, the degree of expression in specific cell types (e.g.
hepatocytes, myeloid cells,
endothelial cells, cancer cells, etc.) can be reduced.
In one embodiment, a microRNA site can be engineered near the 5' terminus of
the 3'UTR,
about halfway between the 5' terminus and 3'terminus of the 3'UTR and/or near
the 3'terminus of the
3'UTR. As a non-limiting example, a microRNA site may be engineered near the
5' terminus of the 3'UTR
and about halfway between the 5' terminus and 3'terminus of the 3'UTR. As
another non-limiting
example, a microRNA site may be engineered near the 3'terminus of the 3'UTR
and about halfway
between the 5' terminus and 3'terminus of the 3'UTR. As yet another non-
limiting example, a microRNA
site may be engineered near the 5' terminus of the 3'UTR and near the 3'
terminus of the 3'UTR.
In another embodiment, a 3'UTR can comprise 4 microRNA sites. The microRNA
sites may
be complete microRNA binding sites, microRNA seed sequences and/or microRNA
binding site
sequences without the seed sequence.
In one embodiment, a nucleic acid of the invention may be engineered to
include at least one
microRNA in order to dampen the antigen presentation by antigen presenting
cells. The microRNA may
be the complete microRNA sequence, the microRNA seed sequence, the microRNA
sequence without
the seed or a combination thereof. As a non-limiting example, the microRNA
incorporated into the nucleic
acid may be specific to the hematopoietic system. As another non-limiting
example, the microRNA
incorporated into the nucleic acid of the invention to dampen antigen
presentation is miR-142-3p.
In one embodiment, a nucleic acid may be engineered to include microRNA sites
which are
expressed in different tissues of a subject. As a non-limiting example, a
modified nucleic acid, enhanced
modified RNA or ribonucleic acid of the present invention may be engineered to
include miR-192 and
miR-122 to regulate expression of the modified nucleic acid, enhanced modified
RNA or ribonucleic acid
in the liver and kidneys of a subject. In another embodiment, a modified
nucleic acid, enhanced modified
RNA or ribonucleic acid may be engineered to include more than one microRNA
sites for the same tissue.
For example, a modified nucleic acid, enhanced modified RNA or ribonucleic
acid of the present invention
may be engineered to include miR-17-92 and miR-126 to regulate expression of
the modified nucleic
acid, enhanced modified RNA or ribonucleic acid in endothelial cells of a
subject.
In one embodiment, the therapeutic window and or differential expression
associated with the
target polypeptide encoded by the modified nucleic acid, enhanced modified RNA
or ribonucleic acid
encoding a signal (also referred to herein as a polynucleotide) of the
invention may be altered. For
example, polynucleotides may be designed whereby a death signal is more highly
expressed in cancer
cells (or a survival signal in a normal cell) by virtue of the miRNA signature
of those cells. Where a cancer
- 162 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
cell expresses a lower level of a particular miRNA, the polynucleotide
encoding the binding site for that
miRNA (or miRNAs) would be more highly expressed. Hence, the target
polypeptide encoded by the
polynucleotide is selected as a protein which triggers or induces cell death.
Neighboring noncancer cells,
harboring a higher expression of the same miRNA would be less affected by the
encoded death signal as
the polynucleotide would be expressed at a lower level due to the effects of
the miRNA binding to the
binding site or "sensor" encoded in the 3'UTR. Conversely, cell survival or
cytoprotective signals may be
delivered to tissues containing cancer and non-cancerous cells where a miRNA
has a higher expression
in the cancer cells-the result being a lower survival signal to the cancer
cell and a larger survival
signature to the normal cell. Multiple polynucleotides may be designed and
administered having different
signals according to the previous paradigm.
In one embodiment, the expression of a nucleic acid may be controlled by
incorporating at
least one sensor sequence in the nucleic acid and formulating the nucleic
acid. As a non-limiting
example, a nucleic acid may be targeted to an orthotopic tumor by having a
nucleic acid incorporating a
miR-122 binding site and formulated in a lipid nanoparticle comprising the
cationic lipid DLin-KC2-DMA.
According to the present invention, the polynucleotides may be modified as to
avoid the
deficiencies of other polypeptide-encoding molecules of the art. Hence, in
this embodiment the
polynucleotides are referred to as modified polynucleotides.
Through an understanding of the expression patterns of microRNA in different
cell types,
modified nucleic acids, enhanced modified RNA or ribonucleic acids such as
polynucleotides can be
engineered for more targeted expression in specific cell types or only under
specific biological conditions.
Through introduction of tissue-specific microRNA binding sites, modified
nucleic acids, enhanced
modified RNA or ribonucleic acids, could be designed that would be optimal for
protein expression in a
tissue or in the context of a biological condition.
Transfection experiments can be conducted in relevant cell lines, using
engineered modified
nucleic acids, enhanced modified RNA or ribonucleic acids and protein
production can be assayed at
various time points post-transfection. For example, cells can be transfected
with different microRNA
binding site-engineering nucleic acids or mRNA and by using an ELISA kit to
the relevant protein and
assaying protein produced at 6 hr, 12 hr, 24 hr, 48 hr, 72 hr and 7 days post-
transfection. In vivo
experiments can also be conducted using microRNA-binding site-engineered
molecules to examine
changes in tissue-specific expression of formulated modified nucleic acids,
enhanced modified RNA or
ribonucleic acids.
Non-limiting examples of cell lines which may be useful in these
investigations include those
from ATCC (Manassas, VA) including MRC-5, A549, T84, NCI-H2126 [H2126], NCI-
H1688 [H1688], WI-
38, WI-38 VA-13 subline 2RA, WI-26 VA4, C3A [HepG2/C3A, derivative of Hep G2
(ATCC HB-8065)],
THLE-3, H69AR, NCI-H292 [H292], CFPAC-1, NTERA-2 cl.D1 [NT2/D1], DMS 79, DMS
53, DMS 153,
DMS 114, MSTO-211H, SW 1573 [SW-1573, SW1573], SW 1271 [SW-1271, 5W1271], SHP-
77, SNU-
398, SNU-449, SNU-182, SNU-475, SNU-387, SNU-423, NL20, NL20-TA [NL20T-A],
THLE-2,
HBE135-E6E7, HCC827, HCC4006, NCI-H23 [H23], NCI-H1299, NCI-H187 [H187], NCI-
H358 [H-358,
H358], NCI-H378 [H378], NCI-H522 [H522], NCI-H526 [H526], NCI-H727 [H727], NCI-
H810 [H810], NCI-
H889 [H889], NCI-H1155 [H1155], NCI-H1404 [H1404], NCI-N87 [N87], NCI-H196
[H196], NCI-H211
[H211], NCI-H220 [H220], NCI-H250 [H250], NCI-H524 [H524], NCI-H647 [H647],
NCI-H650 [H650], NCI-
- 163 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
H711 [H711], NCI-H719 [H719], NCI-H740 [H740], NCI-H748 [H748], NCI-H774
[H774], NCI-H838
[H838], NCI-H841 [H841], NCI-H847 [H847], NCI-H865 [H865], NCI-H920 [H920],
NCI-H1048 [H1048],
NCI-H1092 [H1092], NCI-H1105 [H1105], NCI-H1184 [H1184], NCI-H1238 [H1238],
NCI-H1341 [H1341],
NCI-H1385 [H1385], NCI-H1417 [H1417], NCI-H1435 [H1435], NCI-H1436 [H1436],
NCI-H1437 [H1437],
NCI-H1522 [H1522], NCI-H1563 [H1563], NCI-H1568 [H1568], NCI-H1573 [H1573],
NCI-H1581 [H1581],
NCI-H1618 [H1618], NCI-H1623 [H1623], NCI-H1650 [H-1650, H1650], NCI-H1651
[H1651], NCI-H1666
[H-1666, H1666], NCI-H1672 [H1672], NCI-H1693 [H1693], NCI-H1694 [H1694], NCI-
H1703 [H1703],
NCI-H1734 [H-1734, H1734], NCI-H1755 [H1755], NCI-H1755 [H1755], NCI-H1770
[H1770], NCI-H1793
[H1793], NCI-H1836 [H1836], NCI-H1838 [H1838], NCI-H1869 [H1869], NCI-H1876
[H1876], NCI-H1882
[H1882], NCI-H1915 [H1915], NCI-H1930 [H1930], NCI-H1944 [H1944], NCI-H1975 [H-
1975, H1975],
NCI-H1993 [H1993], NCI-H2023 [H2023], NCI-H2029 [H2029], NCI-H2030 [H2030],
NCI-H2066 [H2066],
NCI-H2073 [H2073], NCI-H2081 [H2081], NCI-H2085 [H2085], NCI-H2087 [H2087],
NCI-H2106 [H2106],
NCI-H2110 [H2110], NCI-H2135 [H2135], NCI-H2141 [H2141], NCI-H2171 [H2171],
NCI-H2172 [H2172],
NCI-H2195 [H2195], NCI-H2196 [H2196], NCI-H2198 [H2198], NCI-H2227 [H2227],
NCI-H2228 [H2228],
NCI-H2286 [H2286], NCI-H2291 [H2291], NCI-H2330 [H2330], NCI-H2342 [H2342],
NCI-H2347 [H2347],
NCI-H2405 [H2405], NCI-H2444 [H2444], UMC-11, NCI-H64 [H64], NCI-H735 [H735],
NCI-H735 [H735],
NCI-H1963 [H1963], NCI-H2107 [H2107], NCI-H2108 [H2108], NCI-H2122 [H2122], Hs
573.T, Hs
573.Lu, PLC/PRF/5, BEAS-2B, Hep G2, Tera-1, Tera-2, NCI-H69 [H69], NCI-H128
[H128], ChaGo-K-
1, NCI-H446 [H446], NCI-H209 [H209], NCI-H146 [H146], NCI-H441 [H441], NCI-H82
[H82], NCI-H460
[H460], NCI-H596 [H596], NCI-H676B [H67613], NCI-H345 [H345], NCI-H820 [H820],
NCI-H520 [H520],
NCI-H661 [H661], NCI-H510A [H510A, NCI-H510], SK-HEP-1, A-427, Calu-1, Calu-3,
Calu-6, SK-LU-
1, SK-MES-1, SW 900 [SW-900, 5W900], MaIme-3M, and Capan-1.
In some embodiments, modified messenger RNA can be designed to incorporate
microRNA
binding region sites that either have 100% identity to known seed sequences or
have less than 100%
identity to seed sequences. The seed sequence can be partially mutated to
decrease microRNA binding
affinity and as such result in reduced downmodulation of that mRNA transcript.
In essence, the degree of
match or mis-match between the target mRNA and the microRNA seed can act as a
rheostat to more
finely tune the ability of the microRNA to modulate protein expression. In
addition, mutation in the non-
seed region of a microRNA binding site may also impact the ability of a
microRNA to modulate protein
expression.
In one embodiment, a miR sequence may be incorporated into the loop of a stem
loop.
In another embodiment, a miR seed sequence may be incorporated in the loop of
a stem loop
and a miR binding site may be incorporated into the 5' or 3' stem of the stem
loop.
In one embodiment, a TEE may be incorporated on the Send of the stem of a stem
loop and a
miR seed may be incorporated into the stem of the stem loop. In another
embodiment, a TEE may be
incorporated on the Send of the stem of a stem loop, a miR seed may be
incorporated into the stem of
the stem loop and a miR binding site may be incorporated into the 3'end of the
stem or the sequence
after the stem loop. The miR seed and the miR binding site may be for the same
and/or different miR
sequences.
In one embodiment, the incorporation of a miR sequence and/or a TEE sequence
changes the
shape of the stem loop region which may increase and/or decrease translation.
(see e.g, Kedde et al. A
- 164 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Pumilio-induced RNA structure switch in p27-3'UTR controls miR-221 and miR-22
accessibility. Nature
Cell Biology. 2010, the sequences of which are incorporated herein by
reference).
In one embodiment, the incorporation of a miR sequence and/or a TEE sequence
changes the
shape of the stem loop region which may increase and/or decrease translation.
(see e.g, Kedde et al. A
Pumilio-induced RNA structure switch in p27-3'UTR controls miR-221 and miR-22
accessibility. Nature
Cell Biology. 2010, the sequences of which are incorporated herein by
reference).
In one embodiment, the 5'UTR may comprise at least one microRNA sequence. The
microRNA sequence may be, but is not limited to, a 19 or 22 nucleotide
sequence and/or a microRNA
sequence without the seed.
In one embodiment the microRNA sequence in the 5'UTR may be used to stabilize
the nucleic
acid and/or mRNA described herein.
In another embodiment, a microRNA sequence in the 5'UTR may be used to
decrease the
accessibility of the site of translation initiation such as, but not limited
to a start codon. Matsuda et al
(PLoS One. 2010 11(5):e15057; the sequences of which are incorporated herein
by reference) used
antisense locked nucleic acid (LNA) oligonucleotides and exon-junction
complexes (EJCs) around a start
codon (-4 to +37 where the A of the AUG codons is +1) in order to decrease the
accessibility to the first
start codon (AUG). Matsuda showed that altering the sequence around the start
codon with an LNA or
EJC the efficiency, length and structural stability of the nucleic acid or
mRNA is affected. The nucleic
acids or mRNA of the present invention may comprise a microRNA sequence,
instead of the LNA or EJC
sequence described by Matsuda et al, near the site of translation initiation
in order to decrease the
accessibility to the site of translation initiation. The site of translation
initiation may be prior to, after or
within the microRNA sequence. As a non-limiting example, the site of
translation initiation may be located
within a microRNA sequence such as a seed sequence or binding site. As another
non-limiting example,
the site of translation initiation may be located within a miR-122 sequence
such as the seed sequence or
the mir-122 binding site.
In one embodiment, the nucleic acids or mRNA of the present invention may
include at least
one microRNA in order to dampen the antigen presentation by antigen presenting
cells. The microRNA
may be the complete microRNA sequence, the microRNA seed sequence, the
microRNA sequence
without the seed or a combination thereof. As a non-limiting example, the
microRNA incorporated into
the nucleic acids or mRNA of the present invention may be specific to the
hematopoietic system. As
another non-limiting example, the microRNA incorporated into the nucleic acids
or mRNA of the present
invention to dampen antigen presentation is miR-142-3p.
In one embodiment, the nucleic acids or mRNA of the present invention may
include at least
one microRNA in order to dampen expression of the encoded polypeptide in a
cell of interest. As a non-
limiting example, the nucleic acids or mRNA of the present invention may
include at least one miR-122
binding site in order to dampen expression of an encoded polypeptide of
interest in the liver. As another
non-limiting example, the nucleic acids or mRNA of the present invention may
include at least one miR-
142-3p binding site, miR-142-3p seed sequence, miR-142-3p binding site without
the seed, miR-142-5p
binding site, miR-142-5p seed sequence, miR-142-5p binding site without the
seed, miR-146 binding site,
miR-146 seed sequence and/or miR-146 binding site without the seed sequence.
- 165 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In one embodiment, the nucleic acids or mRNA of the present invention may
comprise at least
one microRNA binding site in the 3'UTR in order to selectively degrade mRNA
therapeutics in the
immune cells to subdue unwanted immunogenic reactions caused by therapeutic
delivery. As a non-
limiting example, the microRNA binding site may be the modified nucleic acids
more unstable in antigen
presenting cells. Non-limiting examples of these microRNA include mir-142-5p,
mir-142-3p, mir-146a-5p
and mir-146-3p.
In one embodiment, the nucleic acids or mRNA of the present invention
comprises at least
one microRNA sequence in a region of the nucleic acid or mRNA which may
interact with a RNA binding
protein.
RNA Motifs for RNA Binding Proteins (RBPs)
RNA binding proteins (RBPs) can regulate numerous aspects of co- and post-
transcription
gene expression such as, but not limited to, RNA splicing, localization,
translation, turnover,
polyadenylation, capping, modification, export and localization. RNA-binding
domains (RBDs), such as,
but not limited to, RNA recognition motif (RR) and hnRNP K-homology (KH)
domains, typically regulate
the sequence association between RBPs and their RNA targets (Ray et al. Nature
2013. 499:172-177;
the sequences of which are incorporated herein by reference). In one
embodiment, the canonical RBDs
can bind short RNA sequences. In another embodiment, the canonical RBDs can
recognize structure
RNAs.
In one embodiment, to increase the stability of the mRNA of interest, an mRNA
encoding HuR
can be co-transfected or co-injected along with the mRNA of interest into the
cells or into the tissue.
These proteins can also be tethered to the mRNA of interest in vitro and then
administered to the cells
together. Poly A tail binding protein, PABP interacts with eukaryotic
translation initiation factor elF4G to
stimulate translational initiation. Co-administration of mRNAs encoding these
RBPs along with the mRNA
drug and/or tethering these proteins to the mRNA drug in vitro and
administering the protein-bound
mRNA into the cells can increase the translational efficiency of the mRNA. The
same concept can be
extended to co-administration of mRNA along with mRNAs encoding various
translation factors and
facilitators as well as with the proteins themselves to influence RNA
stability and/or translational
efficiency.
In one embodiment, the nucleic acids and/or mRNA may comprise at least one RNA-
binding
motif such as, but not limited to a RNA-binding domain (RBD).
In one embodiment, the RBD may be any of the RBDs, fragments or variants
thereof descried
by Ray et al. (Nature 2013. 499:172-177; the RBDs of which are incorporated
herein by reference).
In one embodiment, the nucleic acids or mRNA of the present invention may
comprise a
sequence for at least one RNA-binding domain (RBDs). When the nucleic acids or
mRNA of the present
invention comprise more than one RBD, the RBDs do not need to be from the same
species or even the
same structural class.
In one embodiment, at least one flanking region (e.g., the 5'UTR and/or the
3'UTR) may
comprise at least one RBD. In another embodiment, the first flanking region
and the second flanking
region may both comprise at least one RBD. The RBD may be the same or each of
the RBDs may have
at least 60% sequence identity to the other RBD. As a non-limiting example, at
least on RBD may be
- 166 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
located before, after and/or within the 3'UTR of the nucleic acid or mRNA of
the present invention. As
another non-limiting example, at least one RBD may be located before or within
the first 300 nucleosides
of the 3'UTR.
In another embodiment, the nucleic acids and/or mRNA of the present invention
may comprise
at least one RBD in the first region of linked nucleosides. The RBD may be
located before, after or within
a coding region (e.g., the ORF).
In yet another embodiment, the first region of linked nucleosides and/or at
least one flanking
region may comprise at least on RBD. As a non-limiting example, the first
region of linked nucleosides
may comprise a RBD related to splicing factors and at least one flanking
region may comprise a RBD for
stability and/or translation factors.
In one embodiment, the nucleic acids and/or mRNA of the present invention may
comprise at
least one RBD located in a coding and/or non-coding region of the nucleic
acids and/or mRNA.
In one embodiment, at least one RBD may be incorporated into at least one
flanking region to
increase the stability of the nucleic acid and/or mRNA of the present
invention.
In one embodiment, a microRNA sequence in a RNA binding protein motif may be
used to
decrease the accessibility of the site of translation initiation such as, but
not limited to a start codon. The
nucleic acids or mRNA of the present invention may comprise a microRNA
sequence, instead of the LNA
or EJC sequence described by Matsuda et al, near the site of translation
initiation in order to decrease the
accessibility to the site of translation initiation. The site of translation
initiation may be prior to, after or
within the microRNA sequence. As a non-limiting example, the site of
translation initiation may be located
within a microRNA sequence such as a seed sequence or binding site. As another
non-limiting example,
the site of translation initiation may be located within a miR-122 sequence
such as the seed sequence or
the mir-122 binding site.
In another embodiment, an antisense locked nucleic acid (LNA) oligonucleotides
and exon-
junction complexes (EJCs) may be used in the RNA binding protein motif. The
LNA and EJCs may be
used around a start codon (-4 to +37 where the A of the AUG codons is +1) in
order to decrease the
accessibility to the first start codon (AUG).
Codon Optimization
The polynucleotides of the invention, their regions or parts or subregions may
be codon
optimized. Codon optimization methods are known in the art and may be useful
in efforts to achieve one
or more of several goals. These goals include to match codon frequencies in
target and host organisms to
ensure proper folding, bias GC content to increase mRNA stability or reduce
secondary structures,
minimize tandem repeat codons or base runs that may impair gene construction
or expression, customize
transcriptional and translational control regions, insert or remove protein
trafficking sequences,
remove/add post translation modification sites in encoded protein (e.g.,
glycosylation sites), add, remove
or shuffle protein domains, insert or delete restriction sites, modify
ribosome binding sites and mRNA
degradation sites, to adjust translational rates to allow the various domains
of the protein to fold properly,
or to reduce or eliminate problem secondary structures within the
polynucleotide. Codon optimization
tools, algorithms and services are known in the art, non-limiting examples
include services from GeneArt
(Life Technologies), DNA2.0 (Menlo Park CA) and/or proprietary methods. In one
embodiment, the ORF
- 167 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
sequence is optimized using optimization algorithms. Codon options for each
amino acid are given in
Table 8.
Table 8: Codon Options.
Amino Acid Single Letter Codon Options
Code
Isoleucine I ATT, ATC, ATA
Leucine L CTT, CTC, CTA, CTG, TTA, TTG
Valine V GTT, GTC, GTA, GTG
Phenylalanine F TTT, TTC
Methionine M ATG
Cysteine C TGT, TGC
Alanine A GCT, GCC, GCA, GCG
Glycine G GGT, GGC, GGA, GGG
Proline P CCT, CCC, CCA, CCG
Threonine T ACT, ACC, ACA, ACG
Serine S TCT, TCC, TCA, TCG, AGT, AGC
Tyrosine Y TAT, TAC
Tryptophan W TGG
Glutamine Q CAA, CAG
Asparagine N AAT, AAC
Histidine H CAT, CAC
Glutamic acid E GAA, GAG
Aspartic acid D GAT, GAC
Lysine K AAA, AAG
Arginine R CGT, CGC, CGA, CGG, AGA, AGG
Selenocysteine Sec UGA in mRNA in presence of
Selenocystein insertion element (SECIS)
Stop codons Stop TAA, TAG, TGA
"Codon optimized" refers to the modification of a starting nucleotide sequence
by replacing at
least one codon of the starting nucleotide sequence with a codon that is more
frequently used in the
group of abundant polypeptides of the host organism. Table 9 contains the
codon usage frequency for
humans (Codon usage database: [[www.]]kazusa.or.jp/codonicgi-
binishowcodon.cgi?species=9606&aa=1&style=N).
Codon optimization may be used to increase the expression of polypeptides by
the
replacement of at least one, at least two, at least three, at least four, at
least five, at least six, at least
seven, at least eight, at least nine, at least ten or at least 1 `)/0, at
least 2%, at least 4%, at least 6%, at
least 8%, at least 10%, at least 20%, at least 40%, at least 60%, at least
80%, at least 90% or at least
95%, or all codons of the starting nucleotide sequence with more frequently or
the most frequently used
codons for the respective amino acid as determined for the group of abundant
proteins.
In one embodiment of the invention, the modified nucleotide sequences contain
for each
amino acid the most frequently used codons of the abundant proteins of the
respective host cell.
- 168 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 9: Codon usage frequency table for humans.
Amino Amino Amino Amino
Codon % Codon % Codon % Codon
%
Acid Acid Acid Acid
UUU F 46 UCU S 19 UAU Y 44 UGU C 46
UUC F 54 UCC S 22 UAC Y 56 UGC C 54
UUA L 8 UCA S 15 UAA * 30 UGA * 47
UUG L 13 UCG S 5 UAG * 24 UGG W 100
CUU L 13 CCU P 29 CAU H 42 CGU R 8
CUC L 20 CCC P 32 CAC H 58 CGC R 18
CUA L 7 CCA P 28 CAA Q 27 CGA R 11
CUG L 40 CCG P 11 CAG Q 73 CGG R 20
AUU I 36 ACU T 25 AAU N 47 AGU S 15
AUC I 47 ACC T 36 AAC N 53 AGC S 24
AUA I 17 ACA T 28 AAA K 43 AGA R 21
AUG M 100 ACG T 11 AAG K 57 AGG R 21
GUU V 18 GCU A 27 GAU D 46 GGU G 16
GUC V 24 GCC A 40 GAC D 54 GGC G 34
GUA V 12 GCA A 23 GAA E 42 GGA G 25
GUG V 46 GCG A 11 GAG E 58 GGG G 25
In one embodiment, after a nucleotide sequence has been codon optimized it may
be further
evaluated for regions containing restriction sites. At least one nucleotide
within the restriction site regions
may be replaced with another nucleotide in order to remove the restriction
site from the sequence but the
replacement of nucleotides does alter the amino acid sequence which is encoded
by the codon optimized
nucleotide sequence.
Features, which may be considered beneficial in some embodiments of the
present invention,
may be encoded by regions of the polynucleotide and such regions may be
upstream (5') or downstream
(3') to a region which encodes a polypeptide. These regions may be
incorporated into the polynucleotide
before and/or after codon optimization of the protein encoding region or open
reading frame (ORF). It is
not required that a polynucleotide contain both a 5' and 3' flanking region.
Examples of such features
include, but are not limited to, untranslated regions (UTRs), Kozak sequences,
an oligo(dT) sequence,
and detectable tags and may include multiple cloning sites which may have Xbal
recognition.
In some embodiments, a 5' UTR and/or a 3' UTR region may be provided as
flanking regions.
Multiple 5' or 3' UTRs may be included in the flanking regions and may be the
same or of different
sequences. Any portion of the flanking regions, including none, may be codon
optimized and any may
independently contain one or more different structural or chemical
modifications, before and/or after
codon optimization.
After optimization (if desired), the polynucleotides components are
reconstituted and
transformed into a vector such as, but not limited to, plasmids, viruses,
cosmids, and artificial
chromosomes. For example, the optimized polynucleotide may be reconstituted
and transformed into
chemically competent E. coli, yeast, neurospora, maize, drosophila, etc. where
high copy plasmid-like or
chromosome structures occur by methods described herein.
- 169 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Uses of Modified Nucleic Acids
Therapeutic Agents
The modified nucleic acids described herein can be used as therapeutic agents.
For
example, a modified nucleic acid described herein can be administered to an
animal or subject,
wherein the modified nucleic acid is translated in vivo to produce a
therapeutic peptide in the animal
or subject. Accordingly, provided herein are compositions, methods, kits, and
reagents for treatment
or prevention of disease or conditions in humans and other mammals. The active
therapeutic agents
of the present disclosure include modified nucleic acids, cells containing
modified nucleic acids or
polypeptides translated from the modified nucleic acids, polypeptides
translated from modified nucleic
acids, cells contacted with cells containing modified nucleic acids or
polypeptides translated from the
modified nucleic acids, tissues containing cells containing modified nucleic
acids and organs
containing tissues containing cells containing modified nucleic acids.
Provided are methods of inducing translation of a synthetic or recombinant
polynucleotide
to produce a polypeptide in a cell population using the modified nucleic acids
described herein. Such
translation can be in vivo, ex vivo, in culture, or in vitro. The cell
population is contacted with an
effective amount of a composition containing a nucleic acid that has at least
one nucleoside
modification, and a translatable region encoding the polypeptide. The
population is contacted under
conditions such that the nucleic acid is localized into one or more cells of
the cell population and the
recombinant polypeptide is translated in the cell from the nucleic acid.
An effective amount of the composition is provided based, at least in part, on
the target
tissue, target cell type, means of administration, physical characteristics of
the nucleic acid (e.g., size,
and extent of modified nucleosides), and other determinants. In general, an
effective amount of the
composition provides efficient protein production in the cell, preferably more
efficient than a
composition containing a corresponding unmodified nucleic acid. Increased
efficiency may be
demonstrated by increased cell transfection (i.e., the percentage of cells
transfected with the nucleic
acid), increased protein translation from the nucleic acid, decreased nucleic
acid degradation (as
demonstrated, e.g., by increased duration of protein translation from a
modified nucleic acid), or
reduced innate immune response of the host cell or improve therapeutic
utility.
Aspects of the present disclosure are directed to methods of inducing in vivo
translation of
a recombinant polypeptide in a mammalian subject in need thereof. Therein, an
effective amount of a
composition containing a nucleic acid that has at least one nucleoside
modification and a translatable
region encoding the polypeptide is administered to the subject using the
delivery methods described
herein. The nucleic acid is provided in an amount and under other conditions
such that the nucleic
acid is localized into a cell or cells of the subject and the recombinant
polypeptide is translated in the
cell from the nucleic acid. The cell in which the nucleic acid is localized,
or the tissue in which the cell
is present, may be targeted with one or more than one rounds of nucleic acid
administration.
Other aspects of the present disclosure relate to transplantation of cells
containing
modified nucleic acids to a mammalian subject. Administration of cells to
mammalian subjects is
known to those of ordinary skill in the art, such as local implantation (e.g.,
topical or subcutaneous
administration), organ delivery or systemic injection (e.g., intravenous
injection or inhalation), as is the
formulation of cells in pharmaceutically acceptable carrier. Compositions
containing modified nucleic
- 170 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
acids are formulated for administration intramuscularly, transarterially,
intraperitoneally, intravenously,
intranasally, subcutaneously, endoscopically, transdermally, or intrathecally.
In some embodiments,
the composition is formulated for extended release.
The subject to whom the therapeutic agent is administered suffers from or is
at risk of
developing a disease, disorder, or deleterious condition. Provided are methods
of identifying,
diagnosing, and classifying subjects on these bases, which may include
clinical diagnosis, biomarker
levels, genome-wide association studies (GWAS), and other methods known in the
art.
In certain embodiments, the administered modified nucleic acid directs
production of one
or more recombinant polypeptides that provide a functional activity which is
substantially absent in the
cell in which the recombinant polypeptide is translated. For example, the
missing functional activity
may be enzymatic, structural, or gene regulatory in nature.
In other embodiments, the administered modified nucleic acid directs
production of one or
more recombinant polypeptides that replace a polypeptide (or multiple
polypeptides) that is
substantially absent in the cell in which the recombinant polypeptide is
translated. Such absence may
be due to genetic mutation of the encoding gene or regulatory pathway thereof.
In other
embodiments, the administered modified nucleic acid directs production of one
or more recombinant
polypeptides to supplement the amount of polypeptide (or multiple
polypeptides) that is present in the
cell in which the recombinant polypeptide is translated. Alternatively, the
recombinant polypeptide
functions to antagonize the activity of an endogenous protein present in, on
the surface of, or
secreted from the cell. Usually, the activity of the endogenous protein is
deleterious to the subject, for
example, due to mutation of the endogenous protein resulting in altered
activity or localization.
Additionally, the recombinant polypeptide antagonizes, directly or indirectly,
the activity of a biological
moiety present in, on the surface of, or secreted from the cell. Examples of
antagonized biological
moieties include lipids (e.g., cholesterol), a lipoprotein (e.g., low density
lipoprotein), a nucleic acid, a
carbohydrate, or a small molecule toxin.
The recombinant proteins described herein are engineered for localization
within the cell,
potentially within a specific compartment such as the nucleus, or are
engineered for secretion from
the cell or translocation to the plasma membrane of the cell.
As described herein, a useful feature of the modified nucleic acids of the
present
disclosure is the capacity to reduce, evade, avoid or eliminate the innate
immune response of a cell to
an exogenous nucleic acid. Provided are methods for performing the titration,
reduction or
elimination of the immune response in a cell or a population of cells. In some
embodiments, the cell
is contacted with a first composition that contains a first dose of a first
exogenous nucleic acid
including a translatable region and at least one nucleoside modification, and
the level of the innate
immune response of the cell to the first exogenous nucleic acid is determined.
Subsequently, the cell
is contacted with a second composition, which includes a second dose of the
first exogenous nucleic
acid, the second dose containing a lesser amount of the first exogenous
nucleic acid as compared to
the first dose. Alternatively, the cell is contacted with a first dose of a
second exogenous nucleic acid.
The second exogenous nucleic acid may contain one or more modified
nucleosides, which may be
the same or different from the first exogenous nucleic acid or, alternatively,
the second exogenous
nucleic acid may not contain modified nucleosides. The steps of contacting the
cell with the first
-171 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
composition and/or the second composition may be repeated one or more times.
Additionally,
efficiency of protein production (e.g., protein translation) in the cell is
optionally determined, and the
cell may be re-transfected with the first and/or second composition repeatedly
until a target protein
production efficiency is achieved.
Therapeutics for diseases and conditions
Provided are methods for treating or preventing a symptom of diseases
characterized by
missing or aberrant protein activity, by replacing the missing protein
activity or overcoming the
aberrant protein activity. Because of the rapid initiation of protein
production following introduction of
modified mRNAs, as compared to viral DNA vectors, the compounds of the present
disclosure are
particularly advantageous in treating acute diseases such as sepsis, stroke,
and myocardial
infarction. Moreover, the lack of transcriptional regulation of the modified
mRNAs of the present
disclosure is advantageous in that accurate titration of protein production is
achievable. Multiple
diseases are characterized by missing (or substantially diminished such that
proper protein function
does not occur) protein activity. Such proteins may not be present, are
present in very low quantities
or are essentially non-functional. The present disclosure provides a method
for treating such
conditions or diseases in a subject by introducing nucleic acid or cell-based
therapeutics containing
the modified nucleic acids provided herein, wherein the modified nucleic acids
encode for a protein
that replaces the protein activity missing from the target cells of the
subject.
Diseases characterized by dysfunctional or aberrant protein activity include,
but not limited
to, cancer and proliferative diseases, genetic diseases (e.g., cystic
fibrosis), autoimmune diseases,
diabetes, neurodegenerative diseases, cardiovascular diseases, and metabolic
diseases. The
present disclosure provides a method for treating such conditions or diseases
in a subject by
introducing nucleic acid or cell-based therapeutics containing the modified
nucleic acids provided
herein, wherein the modified nucleic acids encode for a protein that
antagonizes or otherwise
overcomes the aberrant protein activity present in the cell of the subject.
Specific examples of a dysfunctional protein are the missense or nonsense
mutation
variants of the cystic fibrosis transmembrane conductance regulator (CFTR)
gene, which produce a
dysfunctional or nonfunctional, respectively, protein variant of CFTR protein,
which causes cystic
fibrosis.
Thus, provided are methods of treating cystic fibrosis in a mammalian subject
by
contacting a cell of the subject with a modified nucleic acid having a
translatable region that encodes
a functional CFTR polypeptide, under conditions such that an effective amount
of the CTFR
polypeptide is present in the cell. Preferred target cells are epithelial
cells, such as the lung, and
methods of administration are determined in view of the target tissue; i.e.,
for lung delivery, the RNA
molecules are formulated for administration by inhalation.
In another embodiment, the present disclosure provides a method for treating
hyperlipidemia in a subject, by introducing into a cell population of the
subject with a modified mRNA
molecule encoding Sortilin, a protein recently characterized by genomic
studies, thereby ameliorating
the hyperlipidemia in a subject. The SORT1 gene encodes a trans-Golgi network
(TGN)
transmembrane protein called Sortilin. Genetic studies have shown that one of
five individuals has a
single nucleotide polymorphism, rs12740374, in the 1p13 locus of the SORT1
gene that predisposes
- 172 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
them to having low levels of low-density lipoprotein (LDL) and very-low-
density lipoprotein (VLDL).
Each copy of the minor allele, present in about 30% of people, alters LDL
cholesterol by 8 mg/dL,
while two copies of the minor allele, present in about 5% of the population,
lowers LDL cholesterol 16
mg/dL. Carriers of the minor allele have also been shown to have a 40%
decreased risk of myocardial
infarction. Functional in vivo studies in mice describes that overexpression
of SORT1 in mouse liver
tissue led to significantly lower LDL-cholesterol levels, as much as 80%
lower, and that silencing
SORT1 increased LDL cholesterol approximately 200% (Musunuru K et al. From
noncoding variant to
phenotype via SORT1 at the 1p13 cholesterol locus. Nature 2010; 466: 714-721).
Methods of cellular nucleic acid delivery
Methods of the present disclosure enhance nucleic acid delivery into a cell
population, in
vivo, ex vivo, or in culture. For example, a cell culture containing a
plurality of host cells (e.g.,
eukaryotic cells such as yeast or mammalian cells) is contacted with a
composition that contains an
enhanced nucleic acid having at least one nucleoside modification and,
optionally, a translatable
region. The composition also generally contains a transfection reagent or
other compound that
increases the efficiency of enhanced nucleic acid uptake into the host cells.
The enhanced nucleic
acid exhibits enhanced retention in the cell population, relative to a
corresponding unmodified nucleic
acid. The retention of the enhanced nucleic acid is greater than the retention
of the unmodified
nucleic acid. In some embodiments, it is at least about 50%, 75%, 90%, 95%,
100%, 150%, 200% or
more than 200% greater than the retention of the unmodified nucleic acid. Such
retention advantage
may be achieved by one round of transfection with the enhanced nucleic acid,
or may be obtained
following repeated rounds of transfection.
In some embodiments, the enhanced nucleic acid is delivered to a target cell
population
with one or more additional nucleic acids. Such delivery may be at the same
time, or the enhanced
nucleic acid is delivered prior to delivery of the one or more additional
nucleic acids. The additional
one or more nucleic acids may be modified nucleic acids or unmodified nucleic
acids. It is
understood that the initial presence of the enhanced nucleic acids does not
substantially induce an
innate immune response of the cell population and, moreover, that the innate
immune response will
not be activated by the later presence of the unmodified nucleic acids. In
this regard, the enhanced
nucleic acid may not itself contain a translatable region, if the protein
desired to be present in the
target cell population is translated from the unmodified nucleic acids.
Targeting Moieties
In embodiments of the present disclosure, modified nucleic acids are provided
to express
a protein-binding partner or a receptor on the surface of the cell, which
functions to target the cell to a
specific tissue space or to interact with a specific moiety, either in vivo or
in vitro. Suitable protein-
binding partners include antibodies and functional fragments thereof, scaffold
proteins, or peptides.
Additionally, modified nucleic acids can be employed to direct the synthesis
and extracellular
localization of lipids, carbohydrates, or other biological moieties.
- 173 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Permanent Gene Expression Silencing
A method for epigenetically silencing gene expression in a mammalian subject,
comprising
a nucleic acid where the translatable region encodes a polypeptide or
polypeptides capable of
directing sequence-specific histone H3 methylation to initiate heterochromatin
formation and reduce
gene transcription around specific genes for the purpose of silencing the
gene. For example, a gain-
of-function mutation in the Janus Kinase 2 gene is responsible for the family
of Myeloproliferative
Diseases.
Delivery of a Detectable or Therapeutic Agent to a Biological Target
The modified nucleosides, modified nucleotides, and modified nucleic acids
described
herein can be used in a number of different scenarios in which delivery of a
substance (the "payload")
to a biological target is desired, for example delivery of detectable
substances for detection of the
target, or delivery of a therapeutic agent. Detection methods can include both
imaging in vitro and in
vivo imaging methods, e.g., immunohistochemistry, bioluminescence imaging
(BLI), Magnetic
Resonance Imaging (MRI), positron emission tomography (PET), electron
microscopy, X-ray
computed tomography, Raman imaging, optical coherence tomography, absorption
imaging, thermal
imaging, fluorescence reflectance imaging, fluorescence microscopy,
fluorescence molecular
tomographic imaging, nuclear magnetic resonance imaging, X-ray imaging,
ultrasound imaging,
photoacoustic imaging, lab assays, or in any situation where
tagging/staining/imaging is required.
For example, the modified nucleosides, modified nucleotides, and modified
nucleic acids
described herein can be used in reprogramming induced pluripotent stem cells
(iPS cells), which can
then be used to directly track cells that are transfected compared to total
cells in the cluster. In
another example, a drug that is attached to the modified nucleic acid via a
linker and is fluorescently
labeled can be used to track the drug in vivo, e.g. intracellularly. Other
examples include the use of a
modified nucleic acid in reversible drug delivery into cells.
The modified nucleosides, modified nucleotides, and modified nucleic acids
described
herein can be used in intracellular targeting of a payload, e.g., detectable
or therapeutic agent, to
specific organelle. Exemplary intracellular targets can include the nuclear
localization for advanced
mRNA processing, or a nuclear localization sequence (NLS) linked to the mRNA
containing an
inhibitor.
In addition, the modified nucleosides, modified nucleotides, and modified
nucleic acids
described herein can be used to deliver therapeutic agents to cells or
tissues, e.g., in living animals.
For example, the modified nucleosides, modified nucleotides, and modified
nucleic acids described
herein can be used to deliver highly polar chemotherapeutics agents to kill
cancer cells. The modified
nucleic acids attached to the therapeutic agent through a linker can
facilitate member permeation
allowing the therapeutic agent to travel into a cell to reach an intracellular
target.
In another example, the modified nucleosides, modified nucleotides, and
modified nucleic
acids can be attached to a viral inhibitory peptide (VIP) through a cleavable
linker. The cleavable
linker will release the VIP and dye into the cell. In another example, the
modified nucleosides,
modified nucleotides, and modified nucleic acids can be attached through the
linker to a ADP-
ribosylate, which is responsible for the actions of some bacterial toxins,
such as cholera toxin,
- 174 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
diphtheria toxin, and pertussis toxin. These toxin proteins are ADP-
ribosyltransferases that modify
target proteins in human cells. For example, cholera toxin ADP-ribosylates G
proteins, causing
massive fluid secretion from the lining of the small intestine, resulting in
life-threatening diarrhea.
Pharmaceutical Compositions
The present disclosure provides proteins generated from modified mRNAs.
Pharmaceutical compositions may optionally comprise one or more additional
therapeutically active
substances. In accordance with some embodiments, a method of administering
pharmaceutical
compositions comprising a modified nucleic acid encoding one or more proteins
to be delivered to a
subject in need thereof is provided. In some embodiments, compositions are
administered to
humans. For the purposes of the present disclosure, the phrase "active
ingredient" generally refers to
a protein, protein encoding or protein-containing complex as described herein.
Although the descriptions of pharmaceutical compositions provided herein are
principally
directed to pharmaceutical compositions which are suitable for administration
to humans, it will be
understood by the skilled artisan that such compositions are generally
suitable for administration to
animals of all sorts. Modification of pharmaceutical compositions suitable for
administration to
humans in order to render the compositions suitable for administration to
various animals is well
understood, and the ordinarily skilled veterinary pharmacologist can design
and/or perform such
modification with merely ordinary, if any, experimentation. Subjects to which
administration of the
pharmaceutical compositions is contemplated include, but are not limited to,
humans and/or other
primates; mammals, including commercially relevant mammals such as cattle,
pigs, horses, sheep,
cats, dogs, mice, and/or rats; and/or birds, including commercially relevant
birds such as chickens,
ducks, geese, and/or turkeys.
Formulations of the pharmaceutical compositions described herein may be
prepared by
any method known or hereafter developed in the art of pharmacology. In
general, such preparatory
methods include the step of bringing the active ingredient into association
with an excipient and/or
one or more other accessory ingredients, and then, if necessary and/or
desirable, shaping and/or
packaging the product into a desired single- or multi-dose unit.
A pharmaceutical composition in accordance with the present disclosure may be
prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a
plurality of single unit
doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical
composition
comprising a predetermined amount of the active ingredient. The amount of the
active ingredient is
generally equal to the dosage of the active ingredient which would be
administered to a subject
and/or a convenient fraction of such a dosage such as, for example, one-half
or one-third of such a
dosage.
Relative amounts of the active ingredient, the pharmaceutically acceptable
excipient,
and/or any additional ingredients in a pharmaceutical composition in
accordance with the present
disclosure will vary, depending upon the identity, size, and/or condition of
the subject treated and
further depending upon the route by which the composition is to be
administered. By way of
example, the composition may comprise between 0.1% and 100% (w/w) active
ingredient.
- 175 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Pharmaceutical formulations may additionally comprise a pharmaceutically
acceptable
excipient, which, as used herein, includes any and all solvents, dispersion
media, diluents, or other
liquid vehicles, dispersion or suspension aids, surface active agents,
isotonic agents, thickening or
emulsifying agents, preservatives, solid binders, lubricants and the like, as
suited to the particular
dosage form desired. Remington's The Science and Practice of Pharmacy, 21st
Edition, A. R.
Gennaro (Lippincott, Williams & Wilkins, Baltimore, MD, 2006) discloses
various excipients used in
formulating pharmaceutical compositions and known techniques for the
preparation thereof. Except
insofar as any conventional excipient medium is incompatible with a substance
or its derivatives, such
as by producing any undesirable biological effect or otherwise interacting in
a deleterious manner with
any other component(s) of the pharmaceutical composition, its use is
contemplated to be within the
scope of this present disclosure.
In some embodiments, a pharmaceutically acceptable excipient is at least 95%,
at least
96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some
embodiments, an excipient is
approved for use in humans and for veterinary use. In some embodiments, an
excipient is approved
by United States Food and Drug Administration. In some embodiments, an
excipient is
pharmaceutical grade. In some embodiments, an excipient meets the standards of
the United States
Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British
Pharmacopoeia, and/or the
International Pharmacopoeia.
Pharmaceutically acceptable excipients used in the manufacture of
pharmaceutical
compositions include, but are not limited to, inert diluents, dispersing
and/or granulating agents,
surface active agents and/or emulsifiers, disintegrating agents, binding
agents, preservatives,
buffering agents, lubricating agents, and/or oils. Such excipients may
optionally be included in
pharmaceutical formulations. Excipients such as cocoa butter and suppository
waxes, coloring
agents, coating agents, sweetening, flavoring, and/or perfuming agents can be
present in the
composition, according to the judgment of the formulator.
Exemplary diluents include, but are not limited to, calcium carbonate, sodium
carbonate,
calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen
phosphate, sodium
phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin,
mannitol, sorbitol, inositol,
sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or
combinations thereof.
Exemplary granulating and/or dispersing agents include, but are not limited
to, potato
starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic
acid, guar gum, citrus pulp,
agar, bentonite, cellulose and wood products, natural sponge, cation-exchange
resins, calcium
carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone)
(crospovidone), sodium
carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-
linked sodium
carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized
starch (starch 1500),
microcrystalline starch, water insoluble starch, calcium carboxymethyl
cellulose, magnesium
aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium
compounds, etc., and/or
combinations thereof.
Exemplary surface active agents and/or emulsifiers include, but are not
limited to, natural
emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth,
chondrux, cholesterol,
xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and
lecithin), colloidal clays (e.g.
- 176 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long
chain amino acid
derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl
alcohol, leyl alcohol, triacetin
monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene
glycol monostearate,
polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid,
acrylic acid polymer, and
carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g.
carboxymethylcellulose sodium,
powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose,
hydroxypropyl methylcellulose,
methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan
monolaurate [Tween 20],
polyoxyethylene sorbitan [Tween 60], polyoxyethylene sorbitan monooleate
[Tween 80], sorbitan
monopalmitate [Span 40], sorbitan monostearate [Span 60], sorbitan tristearate
[Span 65], glyceryl
monooleate, sorbitan monooleate [Span 80]), polyoxyethylene esters (e.g.
polyoxyethylene
monostearate [Myrj 45], polyoxyethylene hydrogenated castor oil,
polyethoxylated castor oil,
polyoxymethylene stearate, and Soluton, sucrose fatty acid esters,
polyethylene glycol fatty acid
esters (e.g. Cremophor ), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl
ether [Brij 30]),
poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine
oleate, sodium oleate,
potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl
sulfate, Pluronic F 68,
Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium
chloride, docusate
sodium, etc. and/or combinations thereof.
Exemplary binding agents include, but are not limited to, starch (e.g.
cornstarch and starch
paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses,
lactose, lactitol, mannitol,);
natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish
moss, panwar gum, ghatti
gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose,
ethylcellulose,
hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose,
microcrystalline
cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum
silicate (Veegure), and
larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol;
inorganic calcium salts;
silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations
thereof.
Exemplary preservatives may include, but are not limited to, antioxidants,
chelating
agents, antimicrobial preservatives, antifungal preservatives, alcohol
preservatives, acidic
preservatives, and/or other preservatives. Exemplary antioxidants include, but
are not limited to,
alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole,
butylated
hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid,
propyl gallate, sodium
ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite.
Exemplary chelating agents
include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate,
disodium edetate,
dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid,
sodium edetate, tartaric
acid, and/or trisodium edetate. Exemplary antimicrobial preservatives include,
but are not limited to,
benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol,
cetrimide, cetylpyridinium
chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol,
ethyl alcohol, glycerin,
hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol,
phenylmercuric nitrate, propylene
glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are
not limited to, butyl
paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid,
hydroxybenzoic acid,
potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate,
and/or sorbic acid.
Exemplary alcohol preservatives include, but are not limited to, ethanol,
polyethylene glycol, phenol,
- 177 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or
phenylethyl alcohol.
Exemplary acidic preservatives include, but are not limited to, vitamin A,
vitamin C, vitamin E, beta-
carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic
acid, and/or phytic acid.
Other preservatives include, but are not limited to, tocopherol, tocopherol
acetate, deteroxime
mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened
(BHT),
ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate
(SLES), sodium bisulfite,
sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus
, Phenonip ,
methylparaben, Germall 115, Germaben 11, Neolone¨, Kathon¨, and/or Euxyl .
Exemplary buffering agents include, but are not limited to, citrate buffer
solutions, acetate
buffer solutions, phosphate buffer solutions, ammonium chloride, calcium
carbonate, calcium
chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium
gluconate, d-gluconic acid,
calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate,
pentanoic acid, dibasic
calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium
hydroxide phosphate,
potassium acetate, potassium chloride, potassium gluconate, potassium
mixtures, dibasic potassium
phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium
acetate,
sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic
sodium phosphate,
monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium
hydroxide,
aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline,
Ringer's solution, ethyl alcohol,
etc., and/or combinations thereof.
Exemplary lubricating agents include, but are not limited to, magnesium
stearate, calcium
stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated
vegetable oils, polyethylene
glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium
lauryl sulfate, sodium
lauryl sulfate, etc., and combinations thereof.
Exemplary oils include, but are not limited to, almond, apricot kernel,
avocado, babassu,
bergamot, black current seed, borage, cade, camomile, canola, caraway,
carnauba, castor,
cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu,
eucalyptus, evening
primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop,
isopropyl myristate, jojoba,
kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow,
mango seed,
meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm
kernel, peach kernel,
peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower,
sandalwood,
sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean,
sunflower, tea tree,
thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils
include, but are not limited to,
butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone,
diethyl sebacate, dimethicone
360, isopropyl myristate, mineral oil, octyldodecanol, leyl alcohol, silicone
oil, and/or combinations
thereof.
Liquid dosage forms for oral and parenteral administration include, but are
not limited to,
pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions,
syrups, and/or
elixirs. In addition to active ingredients, liquid dosage forms may comprise
inert diluents commonly
used in the art such as, for example, water or other solvents, solubilizing
agents and emulsifiers such
as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl
alcohol, benzyl benzoate,
propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular,
cottonseed, groundnut,
- 178 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl
alcohol, polyethylene glycols
and fatty acid esters of sorbitan, and mixtures thereof. Besides inert
diluents, oral compositions can
include adjuvants such as wetting agents, emulsifying and suspending agents,
sweetening, flavoring,
and/or perfuming agents. In certain embodiments for parenteral administration,
compositions are
mixed with solubilizing agents such as Cremophor , alcohols, oils, modified
oils, glycols,
polysorbates, cyclodextrins, polymers, and/or combinations thereof.
Injectable preparations, for example, sterile injectable aqueous or oleaginous
suspensions
may be formulated according to the known art using suitable dispersing agents,
wetting agents,
and/or suspending agents. Sterile injectable preparations may be sterile
injectable solutions,
suspensions, and/or emulsions in nontoxic parenterally acceptable diluents
and/or solvents, for
example, as a solution in 1,3-butanediol. Among the acceptable vehicles and
solvents that may be
employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride
solution. Sterile, fixed
oils are conventionally employed as a solvent or suspending medium. For this
purpose any bland
fixed oil can be employed including synthetic mono- or diglycerides. Fatty
acids such as oleic acid
can be used in the preparation of injectables.
Injectable formulations can be sterilized, for example, by filtration through
a bacterial-
retaining filter, and/or by incorporating sterilizing agents in the form of
sterile solid compositions which
can be dissolved or dispersed in sterile water or other sterile injectable
medium prior to use.
In order to prolong the effect of an active ingredient, it is often desirable
to slow the
absorption of the active ingredient from subcutaneous or intramuscular
injection. This may be
accomplished by the use of a liquid suspension of crystalline or amorphous
material with poor water
solubility. The rate of absorption of the drug then depends upon its rate of
dissolution which, in turn,
may depend upon crystal size and crystalline form. Alternatively, delayed
absorption of a parenterally
administered drug form is accomplished by dissolving or suspending the drug in
an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of the drug
in biodegradable
polymers such as polylactide-polyglycolide. Depending upon the ratio of drug
to polymer and the
nature of the particular polymer employed, the rate of drug release can be
controlled. Examples of
other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
Depot injectable
formulations are prepared by entrapping the drug in liposomes or
microemulsions which are
compatible with body tissues.
Compositions for rectal or vaginal administration are typically suppositories
which can be
prepared by mixing compositions with suitable non-irritating excipients such
as cocoa butter,
polyethylene glycol or a suppository wax which are solid at ambient
temperature but liquid at body
temperature and therefore melt in the rectum or vaginal cavity and release the
active ingredient.
Solid dosage forms for oral administration include capsules, tablets, pills,
powders, and
granules. In such solid dosage forms, an active ingredient is mixed with at
least one inert,
pharmaceutically acceptable excipient such as sodium citrate or dicalcium
phosphate and/or fillers or
extenders (e.g. starches, lactose, sucrose, glucose, mannitol, and silicic
acid), binders (e.g.
carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose,
and acacia), humectants
(e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato
or tapioca starch, alginic
acid, certain silicates, and sodium carbonate), solution retarding agents
(e.g. paraffin), absorption
- 179 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
accelerators (e.g. quaternary ammonium compounds), wetting agents (e.g. cetyl
alcohol and glycerol
monostearate), absorbents (e.g. kaolin and bentonite clay), and lubricants
(e.g. talc, calcium stearate,
magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and
mixtures thereof. In the
case of capsules, tablets and pills, the dosage form may comprise buffering
agents.
Solid compositions of a similar type may be employed as fillers in soft and
hard-filled
gelatin capsules using such excipients as lactose or milk sugar as well as
high molecular weight
polyethylene glycols and the like. Solid dosage forms of tablets, dragees,
capsules, pills, and
granules can be prepared with coatings and shells such as enteric coatings and
other coatings well
known in the pharmaceutical formulating art. They may optionally comprise
opacifying agents and
can be of a composition that they release the active ingredient(s) only, or
preferentially, in a certain
part of the intestinal tract, optionally, in a delayed manner. Examples of
embedding compositions
which can be used include polymeric substances and waxes. Solid compositions
of a similar type
may be employed as fillers in soft and hard-filled gelatin capsules using such
excipients as lactose or
milk sugar as well as high molecular weight polyethylene glycols and the like.
Dosage forms for topical and/or transdermal administration of a composition
may include
ointments, pastes, creams, lotions, gels, powders, solutions, sprays,
inhalants and/or patches.
Generally, an active ingredient is admixed under sterile conditions with a
pharmaceutically acceptable
excipient and/or any needed preservatives and/or buffers as may be required.
Additionally, the
present disclosure contemplates the use of transdermal patches, which often
have the added
advantage of providing controlled delivery of a compound to the body. Such
dosage forms may be
prepared, for example, by dissolving and/or dispensing the compound in the
proper medium.
Alternatively or additionally, rate may be controlled by either providing a
rate controlling membrane
and/or by dispersing the compound in a polymer matrix and/or gel.
Suitable devices for use in delivering intradermal pharmaceutical compositions
described
herein include short needle devices such as those described in U.S. Patents
4,886,499; 5,190,521;
5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662.
Intradermal compositions
may be administered by devices which limit the effective penetration length of
a needle into the skin,
such as those described in PCT publication WO 99/34850 and functional
equivalents thereof. Jet
injection devices which deliver liquid compositions to the dermis via a liquid
jet injector and/or via a
needle which pierces the stratum corneum and produces a jet which reaches the
dermis are suitable.
Jet injection devices are described, for example, in U.S. Patents 5,480,381;
5,599,302; 5,334,144;
5,993,412; 5,649,912; 5,569,189; 5,704,911; 5,383,851; 5,893,397; 5,466,220;
5,339,163; 5,312,335;
5,503,627; 5,064,413; 5,520,639; 4,596,556; 4,790,824; 4,941,880; 4,940,460;
and PCT publications
WO 97/37705 and WO 97/13537. Ballistic powder/particle delivery devices which
use compressed
gas to accelerate vaccine in powder form through the outer layers of the skin
to the dermis are
suitable. Alternatively or additionally, conventional syringes may be used in
the classical mantoux
method of intradermal administration.
Formulations suitable for topical administration include, but are not limited
to, liquid and/or
semi liquid preparations such as liniments, lotions, oil in water and/or water
in oil emulsions such as
creams, ointments and/or pastes, and/or solutions and/or suspensions.
Topically-administrable
formulations may, for example, comprise from about 1% to about 10% (w/w)
active ingredient,
- 180 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
although the concentration of active ingredient may be as high as the
solubility limit of the active
ingredient in the solvent. Formulations for topical administration may further
comprise one or more of
the additional ingredients described herein.
A pharmaceutical composition may be prepared, packaged, and/or sold in a
formulation
suitable for pulmonary administration via the buccal cavity. Such a
formulation may comprise dry
particles which comprise the active ingredient and which have a diameter in
the range from about 0.5
nm to about 7 nm or from about 1 nm to about 6 nm. Such compositions are
conveniently in the form
of dry powders for administration using a device comprising a dry powder
reservoir to which a stream
of propellant may be directed to disperse the powder and/or using a self
propelling solvent/powder
dispensing container such as a device comprising the active ingredient
dissolved and/or suspended
in a low-boiling propellant in a sealed container. Such powders comprise
particles wherein at least
98% of the particles by weight have a diameter greater than 0.5 nm and at
least 95% of the particles
by number have a diameter less than 7 nm. Alternatively, at least 95% of the
particles by weight
have a diameter greater than 1 nm and at least 90% of the particles by number
have a diameter less
than 6 nm. Dry powder compositions may include a solid fine powder diluent
such as sugar and are
conveniently provided in a unit dose form.
Low boiling propellants generally include liquid propellants having a boiling
point of below
65 F at atmospheric pressure. Generally the propellant may constitute 50% to
99.9% (w/w) of the
composition, and active ingredient may constitute 0.1% to 20% (w/w) of the
composition. A
propellant may further comprise additional ingredients such as a liquid non-
ionic and/or solid anionic
surfactant and/or a solid diluent (which may have a particle size of the same
order as particles
comprising the active ingredient).
Pharmaceutical compositions formulated for pulmonary delivery may provide an
active
ingredient in the form of droplets of a solution and/or suspension. Such
formulations may be
prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions
and/or suspensions,
optionally sterile, comprising active ingredient, and may conveniently be
administered using any
nebulization and/or atomization device. Such formulations may further comprise
one or more
additional ingredients including, but not limited to, a flavoring agent such
as saccharin sodium, a
volatile oil, a buffering agent, a surface active agent, and/or a preservative
such as
methylhydroxybenzoate. Droplets provided by this route of administration may
have an average
diameter in the range from about 0.1 nm to about 200 nm.
Formulations described herein as being useful for pulmonary delivery are
useful for
intranasal delivery of a pharmaceutical composition. Another formulation
suitable for intranasal
administration is a coarse powder comprising the active ingredient and having
an average particle
from about 0.2 m to 500 m. Such a formulation is administered in the manner
in which snuff is
taken, i.e. by rapid inhalation through the nasal passage from a container of
the powder held close to
the nose.
Formulations suitable for nasal administration may, for example, comprise from
about as
little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may
comprise one or more
of the additional ingredients described herein. A pharmaceutical composition
may be prepared,
-181 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
packaged, and/or sold in a formulation suitable for buccal administration.
Such formulations may, for
example, be in the form of tablets and/or lozenges made using conventional
methods, and may, for
example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally
dissolvable and/or
degradable composition and, optionally, one or more of the additional
ingredients described herein.
Alternately, formulations suitable for buccal administration may comprise a
powder and/or an
aerosolized and/or atomized solution and/or suspension comprising active
ingredient. Such
powdered, aerosolized, and/or aerosolized formulations, when dispersed, may
have an average
particle and/or droplet size in the range from about 0.1 nm to about 200 nm,
and may further
comprise one or more of any additional ingredients described herein.
A pharmaceutical composition may be prepared, packaged, and/or sold in a
formulation
suitable for ophthalmic administration. Such formulations may, for example, be
in the form of eye
drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of
the active ingredient in
an aqueous or oily liquid excipient. Such drops may further comprise buffering
agents, salts, and/or
one or more other of any additional ingredients described herein. Other
opthalmically-administrable
formulations which are useful include those which comprise the active
ingredient in microcrystalline
form and/or in a liposomal preparation. Ear drops and/or eye drops are
contemplated as being within
the scope of this present disclosure.
General considerations in the formulation and/or manufacture of pharmaceutical
agents
may be found, for example, in Remington: The Science and Practice of Pharmacy
21st ed., Lippincott
Williams & Wilkins, 2005 (incorporated herein by reference).
Administration
The present disclosure provides methods comprising administering proteins or
complexes
in accordance with the present disclosure to a subject in need thereof.
Proteins or complexes, or
pharmaceutical, imaging, diagnostic, or prophylactic compositions thereof, may
be administered to a
subject using any amount and any route of administration effective for
preventing, treating,
diagnosing, or imaging a disease, disorder, and/or condition (e.g., a disease,
disorder, and/or
condition relating to working memory deficits). The exact amount required will
vary from subject to
subject, depending on the species, age, and general condition of the subject,
the severity of the
disease, the particular composition, its mode of administration, its mode of
activity, and the like.
Compositions in accordance with the present disclosure are typically
formulated in dosage unit form
for ease of administration and uniformity of dosage. It will be understood,
however, that the total daily
usage of the compositions of the present disclosure will be decided by the
attending physician within
the scope of sound medical judgment. The specific therapeutically effective,
prophylactically
effective, or appropriate imaging dose level for any particular patient will
depend upon a variety of
factors including the disorder being treated and the severity of the disorder;
the activity of the specific
compound employed; the specific composition employed; the age, body weight,
general health, sex
and diet of the patient; the time of administration, route of administration,
and rate of excretion of the
specific compound employed; the duration of the treatment; drugs used in
combination or coincidental
with the specific compound employed; and like factors well known in the
medical arts.
- 182 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or
imaging
compositions thereof may be administered to animals, such as mammals (e.g.,
humans,
domesticated animals, cats, dogs, mice, rats, etc.). In some embodiments,
pharmaceutical,
prophylactic, diagnostic, or imaging compositions thereof are administered to
humans.
Proteins to be delivered and/or pharmaceutical, prophylactic, diagnostic, or
imaging
compositions thereof in accordance with the present disclosure may be
administered by any route. In
some embodiments, proteins and/or pharmaceutical, prophylactic, diagnostic, or
imaging
compositions thereof, are administered by one or more of a variety of routes,
including oral,
intravenous, intramuscular, intra-arterial, intramedullary, intrathecal,
subcutaneous, intraventricular,
transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (e.g.
by powders, ointments,
creams, gels, lotions, and/or drops), mucosa!, nasal, buccal, enteral,
vitreal, intratumoral, sublingual;
by intratracheal instillation, bronchial instillation, and/or inhalation; as
an oral spray, nasal spray,
and/or aerosol, and/or through a portal vein catheter. In some embodiments,
proteins or complexes,
and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions
thereof, are administered by
systemic intravenous injection. In specific embodiments, proteins or complexes
and/or
pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof may
be administered
intravenously and/or orally. In specific embodiments, proteins or complexes,
and/or pharmaceutical,
prophylactic, diagnostic, or imaging compositions thereof, may be administered
in a way which allows
the protein or complex to cross the blood-brain barrier, vascular barrier, or
other epithelial barrier.
However, the present disclosure encompasses the delivery of proteins or
complexes,
and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions
thereof, by any appropriate
route taking into consideration likely advances in the sciences of drug
delivery.
In general the most appropriate route of administration will depend upon a
variety of
factors including the nature of the protein or complex comprising proteins
associated with at least one
agent to be delivered (e.g., its stability in the environment of the
gastrointestinal tract, bloodstream,
etc.), the condition of the patient (e.g., whether the patient is able to
tolerate particular routes of
administration), etc. The present disclosure encompasses the delivery of the
pharmaceutical,
prophylactic, diagnostic, or imaging compositions by any appropriate route
taking into consideration
likely advances in the sciences of drug delivery.
In certain embodiments, compositions in accordance with the present disclosure
may be
administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to
about 100 mg/kg, from
about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg,
from about 0.5 mg/kg
to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1
mg/kg to about 10
mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per
day, one or more times a
day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging
effect. The desired
dosage may be delivered three times a day, two times a day, once a day, every
other day, every third
day, every week, every two weeks, every three weeks, or every four weeks. In
certain embodiments,
the desired dosage may be delivered using multiple administrations (e.g., two,
three, four, five, six,
seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more
administrations).
Proteins or complexes may be used in combination with one or more other
therapeutic,
prophylactic, diagnostic, or imaging agents. By "in combination with," it is
not intended to imply that
- 183 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
the agents must be administered at the same time and/or formulated for
delivery together, although
these methods of delivery are within the scope of the present disclosure.
Compositions can be
administered concurrently with, prior to, or subsequent to, one or more other
desired therapeutics or
medical procedures. In general, each agent will be administered at a dose
and/or on a time schedule
determined for that agent. In some embodiments, the present disclosure
encompasses the delivery
of pharmaceutical, prophylactic, diagnostic, or imaging compositions in
combination with agents that
improve their bioavailability, reduce and/or modify their metabolism, inhibit
their excretion, and/or
modify their distribution within the body.
It will further be appreciated that therapeutically, prophylactically,
diagnostically, or
imaging active agents utilized in combination may be administered together in
a single composition or
administered separately in different compositions. In general, it is expected
that agents utilized in
combination with be utilized at levels that do not exceed the levels at which
they are utilized
individually. In some embodiments, the levels utilized in combination will be
lower than those utilized
individually.
The particular combination of therapies (therapeutics or procedures) to employ
in a
combination regimen will take into account compatibility of the desired
therapeutics and/or
procedures and the desired therapeutic effect to be achieved. It will also be
appreciated that the
therapies employed may achieve a desired effect for the same disorder (for
example, a composition
useful for treating cancer in accordance with the present disclosure may be
administered concurrently
with a chemotherapeutic agent), or they may achieve different effects (e.g.,
control of any adverse
effects).
Kits
The present disclosure provides a variety of kits for conveniently and/or
effectively
carrying out methods of the present disclosure. Typically kits will comprise
sufficient amounts and/or
numbers of components to allow a user to perform multiple treatments of a
subject(s) and/or to
perform multiple experiments.
In one aspect, the disclosure provides kits for protein production, comprising
a first
isolated nucleic acid comprising a translatable region and a nucleic acid
modification, wherein the
nucleic acid is capable of evading or avoiding induction of an innate immune
response of a cell into
which the first isolated nucleic acid is introduced, and packaging and
instructions.
In one aspect, the disclosure provides kits for protein production,
comprising: a first
isolated modified nucleic acid comprising a translatable region, provided in
an amount effective to
produce a desired amount of a protein encoded by the translatable region when
introduced into a
target cell; a second nucleic acid comprising an inhibitory nucleic acid,
provided in an amount
effective to substantially inhibit the innate immune response of the cell; and
packaging and
instructions.
In one aspect, the disclosure provides kits for protein production, comprising
a first
isolated nucleic acid comprising a translatable region and a nucleoside
modification, wherein the
nucleic acid exhibits reduced degradation by a cellular nuclease, and
packaging and instructions.
- 184 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In one aspect, the disclosure provides kits for protein production, comprising
a first
isolated nucleic acid comprising a translatable region and at least two
different nucleoside
modifications, wherein the nucleic acid exhibits reduced degradation by a
cellular nuclease, and
packaging and instructions.
In one aspect, the disclosure provides kits for protein production, comprising
a first
isolated nucleic acid comprising a translatable region and at least one
nucleoside modification,
wherein the nucleic acid exhibits reduced degradation by a cellular nuclease;
a second nucleic acid
comprising an inhibitory nucleic acid; and packaging and instructions.
In some embodiments, the first isolated nucleic acid comprises messenger RNA
(mRNA).
In some embodiments the mRNA comprises at least one nucleoside selected from
the group
consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-
uridine, 2-thiouridine, 4-thio-
pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-
carboxymethyl-uridine, 1-
carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-
taurinomethyluridine,
1-taurinomethyl-pseudouridine, 5-taurinomethy1-2-thio-uridine, 1-taurinomethy1-
4-thio-uridine, 5-
methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-
1-methyl-
pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-
pseudouridine,
dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-
dihydropseudouridine, 2-
methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-
thio-pseudouridine
or any disclosed herein.
In some embodiments, the mRNA comprises at least one nucleoside selected from
the
group consisting of 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-
acetylcytidine, 5-
formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-
pseudoisocytidine, pyrrolo-
cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-
cytidine, 4-thio-pseudoisocytidine,
4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine,
1-methyl-1-deaza-
pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-
thio-zebularine, 2-thio-
zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-
pseudoisocytidine, 4-
methoxy-1-methyl-pseudoisocytidine or any disclosed herein.
In some embodiments, the mRNA comprises at least one nucleoside selected from
the
group consisting of 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-
deaza-8-aza-adenine, 7-
deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-
deaza-8-aza-2,6-
diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine,
N6-(cis-
hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl)
adenosine, N6-
glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-
threonyl
carbamoyladenosine, N6,N6-dimethyladenosine, 7-methyladenine, 2-methylthio-
adenine, 2-methoxy-
adenine or any disclosed herein.
In some embodiments, the mRNA comprises at least one nucleoside selected from
the
group consisting of inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-
guanosine, 7-deaza-8-
aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-
aza-guanosine, 7-
methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-
guanosine, 1-
methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine,
7-methy1-8-oxo-
- 185 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, N2,N2-
dimethy1-6-thio-guanosine
or any disclosed herein.
In another aspect, the disclosure provides compositions for protein
production, comprising
a first isolated nucleic acid comprising a translatable region and a
nucleoside modification, wherein
the nucleic acid exhibits reduced degradation by a cellular nuclease, and a
mammalian cell suitable
for translation of the translatable region of the first nucleic acid.
Definitions
At various places in the present specification, substituents of compounds of
the present
disclosure are disclosed in groups or in ranges. It is specifically intended
that the present disclosure
include each and every individual subcombination of the members of such groups
and ranges. For
example, the term "C1_6 alkyl" is specifically intended to individually
disclose methyl, ethyl, C3 alkyl, C4
alkyl, C5 alkyl, and C6 alkyl.
About: As used herein, the term "about" means +1- 10% of the recited value.
Administered in combination: As used herein, the term "administered in
combination" or
"combined administration" means that two or more agents are administered to a
subject at the same
time or within an interval such that there may be an overlap of an effect of
each agent on the patient.
In some embodiments, they are administered within about 60, 30, 15, 10, 5, or
1 minute of one
another. In some embodiments, the administrations of the agents are spaced
sufficiently closely
together such that a combinatorial (e.g., a synergistic) effect is achieved.
Animal: As used herein, the term "animal" refers to any member of the animal
kingdom.
In some embodiments, "animal" refers to humans at any stage of development. In
some
embodiments, "animal" refers to non-human animals at any stage of development.
In certain
embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat,
a rabbit, a monkey,
a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments,
animals include, but are not
limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some
embodiments, the animal
is a transgenic animal, genetically-engineered animal, or a clone.
Antigens of interest or desired antigens: As used herein, the terms "antigens
of interest" or
"desired antigens" include those proteins and other biomolecules provided
herein that are
immunospecifically bound by the antibodies and fragments, mutants, variants,
and alterations thereof
described herein. Examples of antigens of interest include, but are not
limited to, insulin, insulin-like
growth factor, hGH, tPA, cytokines, such as interleukins (IL), e.g., IL-1, IL-
2, IL-3, IL-4, IL-5, IL-6, IL-7,
IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18,
interferon (IFN) alpha, IFN beta,
IFN gamma, IFN omega or IFN tau, tumor necrosis factor (TNF), such as TNF
alpha and TNF beta,
TNF gamma, TRAIL; G-CSF, GM-CSF, M-CSF, MCP-1 and VEGF.
Approximately: As used herein, the term "approximately" or "about," as applied
to one or
more values of interest, refers to a value that is similar to a stated
reference value. In certain
embodiments, the term "approximately" or "about" refers to a range of values
that fall within 25%,
20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%,
3%, 2%,
1%, or less in either direction (greater than or less than) of the stated
reference value unless
- 186 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
otherwise stated or otherwise evident from the context (except where such
number would exceed
100% of a possible value).
Associated with: As used herein, the terms "associated with," "conjugated,"
"linked,"
"attached," and "tethered," when used with respect to two or more moieties,
means that the moieties
are physically associated or connected with one another, either directly or
via one or more additional
moieties that serves as a linking agent, to form a structure that is
sufficiently stable so that the
moieties remain physically associated under the conditions in which the
structure is used, e.g.,
physiological conditions. An "association" need not be strictly through direct
covalent chemical
bonding. It may also suggest ionic or hydrogen bonding or a hybridization
based connectivity
sufficiently stable such that the "associated" entities remain physically
associated.
Biocompatible: As used herein, the term "biocompatible" means compatible with
living
cells, tissues, organs or systems posing little to no risk of injury, toxicity
or rejection by the immune
system.
Biodegradable: As used herein, the term "biodegradable" means capable of being
broken
down into innocuous products by the action of living things.
Biologically active: As used herein, the phrase "biologically active" refers
to a
characteristic of any substance that has activity in a biological system
and/or organism. For instance,
a substance that, when administered to an organism, has a biological effect on
that organism, is
considered to be biologically active. In particular embodiments, a
polynucleotide of the present
invention may be considered biologically active if even a portion of the
polynucleotide is biologically
active or mimics an activity considered biologically relevant.
Chemical terms: The following provides the definition of various chemical
terms from
"acyl" to "thiol."
The term "acyl," as used herein, represents a hydrogen or an alkyl group
(e.g., a haloalkyl
group), as defined herein, that is attached to the parent molecular group
through a carbonyl group, as
defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group),
acetyl, trifluoroacetyl,
propionyl, butanoyl and the like. Exemplary unsubstituted acyl groups include
from 1 to 7, from 1 to
11, or from 1 to 21 carbons. In some embodiments, the alkyl group is further
substituted with 1, 2, 3,
or 4 substituents as described herein.
The term "acylamino," as used herein, represents an acyl group, as defined
herein,
attached to the parent molecular group though an amino group, as defined
herein (i.e., _N(RN1)C(0)R, where R is H or an optionally substituted C1_6,
C1_10, or C1_20 alkyl group (e.g., haloalkyl) and RN1 is
as defined herein). Exemplary unsubstituted acylamino groups include from 1 to
41 carbons (e.g.,
from 1 to 7, from 1 to 13, from 1 to 21, from 2 to 7, from 2 to 13, from 2 to
21, or from 2 to 41
carbons). In some embodiments, the alkyl group is further substituted with 1,
2, 3, or 4 substituents
as described herein, and/or the amino group is ¨NH2 or ¨NHRN1, wherein RN1 is,
independently, OH,
NO2, NH2, NRN22, SO2ORN2, SO2RN2, SORN2, alkyl, aryl, acyl (e.g., acetyl,
trifluoroacetyl, or others
described herein), or alkoxycarbonylalkyl, and each RN2can be H, alkyl, or
aryl.
The term "acylaminoalkyl," as used herein, represents an acyl group, as
defined herein,
attached to an amino group that is in turn attached to the parent molecular
group though an alkyl
group, as defined herein (i.e., ¨alkyl-N(R)-C(0)-R, where R is H or an
optionally substituted C1_6, C1_
- 187 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
10, or C1-20 alkyl group (e.g., haloalkyl) and RN1 is as defined herein).
Exemplary unsubstituted
acylamino groups include from 1 to 41 carbons (e.g., from 1 to 7, from 1 to
13, from 1 to 21, from 2 to
7, from 2 to 13, from 2 to 21, or from 2 to 41 carbons). In some embodiments,
the alkyl group is
further substituted with 1, 2, 3, or 4 substituents as described herein,
and/or the amino group is -NH2
or -NHRN1, wherein RN1 is, independently, OH, NO2, NH2, NRN22, SO2ORN2,
SO2RN2, SORN2, alkyl,
aryl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), or
alkoxycarbonylalkyl, and each
RN2 can be H, alkyl, or aryl.
The term "acyloxy," as used herein, represents an acyl group, as defined
herein, attached
to the parent molecular group though an oxygen atom (i.e., -0-C(0)-R, where R
is H or an optionally
substituted C1_6, C1-10, or C1_20 alkyl group). Exemplary unsubstituted
acyloxy groups include from 1 to
21 carbons (e.g., from 1 to 7 or from 1 to 11 carbons). In some embodiments,
the alkyl group is
further substituted with 1, 2, 3, or 4 substituents as described herein.
The term "acyloxyalkyl," as used herein, represents an acyl group, as defined
herein,
attached to an oxygen atom that in turn is attached to the parent molecular
group though an alkyl
group (i.e., -alkyl-O-C(0)-R, where R is H or an optionally substituted C1_6,
C1_10, or C1_20 alkyl group).
Exemplary unsubstituted acyloxyalkyl groups include from 1 to 21 carbons
(e.g., from 1 to 7 or from 1
to 11 carbons). In some embodiments, the alkyl group is, independently,
further substituted with 1, 2,
3, or 4 substituents as described herein.
The term "alkaryl," as used herein, represents an aryl group, as defined
herein, attached
to the parent molecular group through an alkylene group, as defined herein.
Exemplary unsubstituted
alkaryl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20
carbons, such as C1_6 alk-
C6_10 aryl, C1_10 alk-C6_10 aryl, or C1_20 alk-C6_10 aryl). In some
embodiments, the alkylene and the aryl
each can be further substituted with 1, 2, 3, or 4 substituent groups as
defined herein for the
respective groups. Other groups preceded by the prefix "alk-" are defined in
the same manner, where
"alk" refers to a C1_6 alkylene, unless otherwise noted, and the attached
chemical structure is as
defined herein.
The term "alkcycloalkyl" represents a cycloalkyl group, as defined herein,
attached to the
parent molecular group through an alkylene group, as defined herein (e.g., an
alkylene group of from
1 to 4, from 1 to 6, from 1 to 10, or form 1 to 20 carbons). In some
embodiments, the alkylene and
the cycloalkyl each can be further substituted with 1, 2, 3, or 4 substituent
groups as defined herein
for the respective group.
The term "alkenyl," as used herein, represents monovalent straight or branched
chain
groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6
or from 2 to 10 carbons)
containing one or more carbon-carbon double bonds and is exemplified by
ethenyl, 1-propenyl, 2-
propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, and the like. Alkenyls
include both cis and trans
isomers. Alkenyl groups may be optionally substituted with 1, 2, 3, or 4
substituent groups that are
selected, independently, from amino, aryl, cycloalkyl, or heterocyclyl (e.g.,
heteroaryl), as defined
herein, or any of the exemplary alkyl substituent groups described herein.
The term "alkenyloxy" represents a chemical substituent of formula -OR, where
R is a C2_
20 alkenyl group (e.g., C2_6 or C2_10 alkenyl), unless otherwise specified.
Exemplary alkenyloxy groups
- 188 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
include ethenyloxy, propenyloxy, and the like. In some embodiments, the
alkenyl group can be
further substituted with 1, 2, 3, or 4 substituent groups as defined herein
(e.g., a hydroxy group).
The term "alkheteroaryl" refers to a heteroaryl group, as defined herein,
attached to the
parent molecular group through an alkylene group, as defined herein. Exemplary
unsubstituted
alkheteroaryl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to
18, from 2 to 17, from 2 to
16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as
C1_6 alk-C1_12 heteroaryl,
C1_10 alk-Ci_12 heteroaryl, or C1_20 alk-C1_12 heteroaryl). In some
embodiments, the alkylene and the
heteroaryl each can be further substituted with 1, 2, 3, or 4 substituent
groups as defined herein for
the respective group. Alkheteroaryl groups are a subset of alkheterocyclyl
groups.
The term "alkheterocyclyl" represents a heterocyclyl group, as defined herein,
attached to
the parent molecular group through an alkylene group, as defined herein.
Exemplary unsubstituted
alkheterocyclyl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to
18, from 2 to 17, from 2
to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such
as C1_6 alk-C1_12
heterocyclyl, C1_10 alk-Ci_12 heterocyclyl, or C1_20 alk-C1_12 heterocyclyl).
In some embodiments, the
alkylene and the heterocyclyl each can be further substituted with 1, 2, 3, or
4 substituent groups as
defined herein for the respective group.
The term "alkoxy" represents a chemical substituent of formula -OR, where R is
a C1_20
alkyl group (e.g., C1_6 or C1_10 alkyl), unless otherwise specified. Exemplary
alkoxy groups include
methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the
like. In some
embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4
substituent groups as
defined herein (e.g., hydroxy or alkoxy).
The term "alkoxyalkoxy" represents an alkoxy group that is substituted with an
alkoxy
group. Exemplary unsubstituted alkoxyalkoxy groups include between 2 to 40
carbons (e.g., from 2
to 12 or from 2 to 20 carbons, such as C1_6 alkoxy-C1_6 alkoxy, C1_10 alkoxy-
C1_10 alkoxy, or C1_20
alkoxy-C1_20 alkoxy). In some embodiments, the each alkoxy group can be
further substituted with 1,
2, 3, or 4 substituent groups as defined herein.
The term "alkoxyalkyl" represents an alkyl group that is substituted with an
alkoxy group.
Exemplary unsubstituted alkoxyalkyl groups include between 2 to 40 carbons
(e.g., from 2 to 12 or
from 2 to 20 carbons, such as C1_6 alkoxy-C1_6 alkyl, C1_10 alkoxy-C1_10
alkyl, or C1_20 alkoxy-C1_20 alkyl).
In some embodiments, the alkyl and the alkoxy each can be further substituted
with 1, 2, 3, or 4
substituent groups as defined herein for the respective group.
The term "alkoxycarbonyl," as used herein, represents an alkoxy, as defined
herein,
attached to the parent molecular group through a carbonyl atom (e.g., -C(0)-
OR, where R is H or an
optionally substituted C1_6, C1-10, or C1_20 alkyl group). Exemplary
unsubstituted alkoxycarbonyl
include from 1 to 21 carbons (e.g., from 1 to 11 or from 1 to 7 carbons). In
some embodiments, the
alkoxy group is further substituted with 1, 2, 3, or 4 substituents as
described herein.
The term "alkoxycarbonylacyl," as used herein, represents an acyl group, as
defined
herein, that is substituted with an alkoxycarbonyl group, as defined herein
(e.g., -C(0) -alkyl-C(0)-
0R, where R is an optionally substituted C1_6, C1_10, or C1_20 alkyl group).
Exemplary unsubstituted
alkoxycarbonylacyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to
13, from 3 to 17, from 3
to 21, or from 3 to 31 carbons, such as C1_6 alkoxycarbonyl-C1_6 acyl, C1_10
alkoxycarbonyl-C1_10 acyl,
- 189 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
or C1_20 alkoxycarbonyl-C1_20 acyl). In some embodiments, each alkoxy and
alkyl group is further
independently substituted with 1, 2, 3, or 4 substituents, as described herein
(e.g., a hydroxy group)
for each group.
The term "alkoxycarbonylalkoxy," as used herein, represents an alkoxy group,
as defined
herein, that is substituted with an alkoxycarbonyl group, as defined herein
(e.g., -0-alkyl-C(0)-0R,
where R is an optionally substituted C1_6, C1_10, or C1_20 alkyl group).
Exemplary unsubstituted
alkoxycarbonylalkoxy include from 3 to 41 carbons (e.g., from 3 to 10, from 3
to 13, from 3 to 17, from
3 to 21, or from 3 to 31 carbons, such as C1_6 alkoxycarbonyl-C1_6 alkoxy,
C1_10 alkoxycarbonyl-C1_10
alkoxy, or C1_20 alkoxycarbonyl-C1_20 alkoxy). In some embodiments, each
alkoxy group is further
independently substituted with 1, 2, 3, or 4 substituents, as described herein
(e.g., a hydroxy group).
The term "alkoxycarbonylalkyl," as used herein, represents an alkyl group, as
defined
herein, that is substituted with an alkoxycarbonyl group, as defined herein
(e.g., -alkyl-C(0)-0R,
where R is an optionally substituted C1-20, C1-10, or C1_6 alkyl group).
Exemplary unsubstituted
alkoxycarbonylalkyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3
to 13, from 3 to 17, from 3
to 21, or from 3 to 31 carbons, such as C1_6 alkoxycarbonyl-C1_6 alkyl, C1_10
alkoxycarbonyl-C1_10 alkyl,
or C1_20 alkoxycarbonyl-C1_20 alkyl). In some embodiments, each alkyl and
alkoxy group is further
independently substituted with 1, 2, 3, or 4 substituents as described herein
(e.g., a hydroxy group).
The term "alkoxycarbonylalkenyl," as used herein, represents an alkenyl group,
as defined
herein, that is substituted with an alkoxycarbonyl group, as defined herein
(e.g., -alkenyl-C(0)-0R,
where R is an optionally substituted C1-20, C1-10, or C1_6 alkyl group).
Exemplary unsubstituted
alkoxycarbonylalkenyl include from 4 to 41 carbons (e.g., from 4 to 10, from 4
to 13, from 4 to 17,
from 4 to 21, or from 4 to 31 carbons, such as C1_6 alkoxycarbonyl-C2_6
alkenyl, C1_10 alkoxycarbonyl-
C2_10 alkenyl, or C1_20 alkoxycarbonyl-C2_20 alkenyl). In some embodiments,
each alkyl, alkenyl, and
alkoxy group is further independently substituted with 1, 2, 3, or 4
substituents as described herein
(e.g., a hydroxy group).
The term "alkoxycarbonylalkynyl," as used herein, represents an alkynyl group,
as defined
herein, that is substituted with an alkoxycarbonyl group, as defined herein
(e.g., -alkynyl-C(0)-0R,
where R is an optionally substituted C1-20, C1-10, or C1_6 alkyl group).
Exemplary unsubstituted
alkoxycarbonylalkynyl include from 4 to 41 carbons (e.g., from 4 to 10, from 4
to 13, from 4 to 17,
from 4 to 21, or from 4 to 31 carbons, such as C1_6 alkoxycarbonyl-C2_6
alkynyl, C1_10 alkoxycarbonyl-
C2_10 alkynyl, or C1_20 alkoxycarbonyl-C2_20 alkynyl). In some embodiments,
each alkyl, alkynyl, and
alkoxy group is further independently substituted with 1, 2, 3, or 4
substituents as described herein
(e.g., a hydroxy group).
The term "alkyl," as used herein, is inclusive of both straight chain and
branched chain
saturated groups from 1 to 20 carbons (e.g., from 1 to 10 or from 1 to 6),
unless otherwise specified.
Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, n-, sec-,
iso- and tert-butyl,
neopentyl, and the like, and may be optionally substituted with one, two,
three, or, in the case of alkyl
groups of two carbons or more, four substituents independently selected from
the group consisting of:
(1) C1_6 alkoxy; (2) C1_6 alkylsulfinyl; (3) amino, as defined herein (e.g.,
unsubstituted amino (i.e., -
NH2) or a substituted amino (i.e., -N(RN1)2, where RN1 is as defined for
amino); (4) C6_10 aryl-C1_6
alkoxy; (5) azido; (6) halo; (7) (C2_9 heterocyclypoxy; (8) hydroxy,
optionally substituted with an 0-
- 190 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
protecting group; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11)
C17 spirocyclyl; (12)
thioalkoxy; (13) thiol; (14) -CO2RA, optionally substituted with an 0-
protecting group and where RA is
selected from the group consisting of (a) C1_20 alkyl (e.g., C1_6 alkyl), (b)
C2_20 alkenyl (e.g., C2-6
alkenyl), (c) C6_10 aryl, (d) hydrogen, (e) C1_6 alk-C6_10 aryl, (f) amino-
C1_20 alkyl, (g) polyethylene glycol
of -(C1-12)s2(OCH2C1-12)si (C1-12)s3OR', wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to
4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0
to 4, from 0 to 6, from 1
to 4, from 1 to 6, or from 1 to 10), and R' is H or C1_20 alkyl, and (h) amino-
polyethylene glycol of -
NRNi(C1-12)s2(CH2CH20)si(C1-12)s3NRN1, wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from
1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently,
hydrogen or optionally
substituted C1_6 alkyl; (15) -C(0)NRB Rc , where each of RB and IR is,
independently, selected from
the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl, and (d)
C1_6 alk-C6_10 aryl; (16) -
SO2RD, where RD is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) C1_6 alk-C6_10
aryl, and (d) hydroxy; (17) -SO2NRERF, where each of RE and RF is,
independently, selected from
the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl and (d)
C1_6 alk-C6_10 aryl; (18) -
C(0)RG , where RG is selected from the group consisting of (a) C1_20 alkyl
(e.g., C1_6 alkyl), (b) C2-20
alkenyl (e.g., C2_6 alkenyl), (c) C6_10 aryl, (d) hydrogen, (e) C1_6 alk-C6_10
aryl, (f) amino-C1_20 alkyl, (g)
polyethylene glycol of -(C1-12)s2(OCH2C1-12)si(C1-12)s3OR', wherein 51 is an
integer from 1 to 10 (e.g.,
from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer
from 0 to 10 (e.g., from 0 to
4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or
C1_20 alkyl, and (h) amino-
polyethylene glycol of -NRN1 (C1-12)s2(CH2CH20)si (C1-12)s3NRN1, wherein 51 is
an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an
integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each
RN1 is, independently,
hydrogen or optionally substituted C1_6 alkyl; (19) -NRHC(0)R1, wherein RH is
selected from the group
consisting of (al) hydrogen and (bl) C1_6 alkyl, and RI is selected from the
group consisting of (a2)
C1_20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2_6 alkenyl), (c2)
C6_10 aryl, (d2) hydrogen, (e2) C1-
6 alk-C6_10 aryl, (f2) amino-C1_20 alkyl, (g2) polyethylene glycol of -
(CH2)s2(OCH2CH2)s1(CH2)s3OR',
wherein 51 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each
of s2 and s3,
independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or
from 1 to 10), and R' is H or C1_20 alkyl, and (h2) amino-polyethylene glycol
of -
NRNi(C1-12)s2(CH2CH20)si (C1-12)s3NRN1, wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from
1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently,
hydrogen or optionally
substituted C1_6 alkyl; (20) -NRJ C(0)ORK , wherein RJ is selected from the
group consisting of (al)
hydrogen and (bl) C1_6 alkyl, and RK is selected from the group consisting of
(a2) C1_20 alkyl (e.g., C1_6
alkyl), (b2) C2_20 alkenyl (e.g., C2_6 alkenyl), (c2) C6_10 aryl, (d2)
hydrogen, (e2) C1_6 alk-C6_10 aryl, (f2)
amino-C1_20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s3OR',
wherein sl is an
integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer
from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or
from 1 to 10), and R' is H or C1_
20 alkyl, and (h2) amino-polyethylene glycol of -
NRN1(CH2)s2(CH2CH20)si(CH2)s3NRN1, wherein sl is
an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an
-191 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to
6, or from 1 to 10), and each
-N1
h< is, independently, hydrogen or optionally substituted C1_6 alkyl; and
(21) amidine. In some
embodiments, each of these groups can be further substituted as described
herein. For example, the
alkylene group of a C1-alkaryl can be further substituted with an oxo group to
afford the respective
aryloyl substituent.
The term "alkylene" and the prefix "alk-," as used herein, represent a
saturated divalent
hydrocarbon group derived from a straight or branched chain saturated
hydrocarbon by the removal
of two hydrogen atoms, and is exemplified by methylene, ethylene,
isopropylene, and the like. The
term "Cx_y alkylene" and the prefix "Cx_y alk-" represent alkylene groups
having between x and y
carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values
for y are 2, 3, 4, 5, 6,
7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., C1_6, C1-10, C2-20, C2-6, C2-10, or
C2_20 alkylene). In some
embodiments, the alkylene can be further substituted with 1, 2, 3, or 4
substituent groups as defined
herein for an alkyl group.
The term "alkylsulfinyl," as used herein, represents an alkyl group attached
to the parent
molecular group through an -S(0)- group. Exemplary unsubstituted alkylsulfinyl
groups are from 1 to
6, from 1 to 10, or from 1 to 20 carbons. In some embodiments, the alkyl group
can be further
substituted with 1, 2, 3, or 4 substituent groups as defined herein.
The term "alkylsulfinylalkyl," as used herein, represents an alkyl group, as
defined herein,
substituted by an alkylsulfinyl group. Exemplary unsubstituted
alkylsulfinylalkyl groups are from 2 to
12, from 2 to 20, or from 2 to 40 carbons. In some embodiments, each alkyl
group can be further
substituted with 1, 2, 3, or 4 substituent groups as defined herein.
The term "alkynyl," as used herein, represents monovalent straight or branched
chain
groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to
10 carbons) containing a
carbon-carbon triple bond and is exemplified by ethynyl, 1-propynyl, and the
like. Alkynyl groups may
be optionally substituted with 1, 2, 3, or 4 substituent groups that are
selected, independently, from
aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or
any of the exemplary alkyl
substituent groups described herein.
The term "alkynyloxy" represents a chemical substituent of formula -OR, where
R is a C2_
20 alkynyl group (e.g., C2_6 or C2_10 alkynyl), unless otherwise specified.
Exemplary alkynyloxy groups
include ethynyloxy, propynyloxy, and the like. In some embodiments, the
alkynyl group can be further
substituted with 1, 2, 3, or 4 substituent groups as defined herein (e.g., a
hydroxy group).
The term "amidine," as used herein, represents a -C(=NH)NH2 group.
The term "amino," as used herein, represents -N(RN1)2, wherein each RN1 is,
independently, H, OH, NO2, N(RN2)2, SO2ORN2, SO2RN2, SORN2, an N-protecting
group, alkyl, alkenyl,
alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, carboxyalkyl (e.g.,
optionally substituted with an
0-protecting group, such as optionally substituted arylalkoxycarbonyl groups
or any described
herein), sulfoalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described
herein), alkoxycarbonylalkyl
(e.g., optionally substituted with an 0-protecting group, such as optionally
substituted
arylalkoxycarbonyl groups or any described herein), heterocyclyl (e.g.,
heteroaryl), or alkheterocyclyl
(e.g., alkheteroaryl), wherein each of these recited RN1 groups can be
optionally substituted, as
defined herein for each group; or two RN1 combine to form a heterocyclyl or an
N-protecting group,
- 192 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
and wherein each RN2 is, independently, H, alkyl, or aryl. The amino groups of
the invention can be
an unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2).
In a preferred
embodiment, amino is -NH2 or -NHRN1, wherein RN1 is, independently, OH, NO2,
NH2, NRN22,
SO2ORN2, SO2RN2, SORN2, alkyl, carboxyalkyl, sulfoalkyl, acyl (e.g., acetyl,
trifluoroacetyl, or others
described herein), alkoxycarbonylalkyl (e.g., t-butoxycarbonylalkyl) or aryl,
and each RN2 can be H,
C1_20 alkyl (e.g., C1_6 alkyl), or C6_10 aryl.
The term "amino acid," as described herein, refers to a molecule having a side
chain, an
amino group, and an acid group (e.g., a carboxy group of -CO2H or a sulfo
group of -S03H), wherein
the amino acid is attached to the parent molecular group by the side chain,
amino group, or acid
group (e.g., the side chain). In some embodiments, the amino acid is attached
to the parent
molecular group by a carbonyl group, where the side chain or amino group is
attached to the carbonyl
group. Exemplary side chains include an optionally substituted alkyl, aryl,
heterocyclyl, alkaryl,
alkheterocyclyl, aminoalkyl, carbamoylalkyl, and carboxyalkyl. Exemplary amino
acids include
alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid,
glutamine, glycine, histidine,
hydroxynorvaline, isoleucine, leucine, lysine, methionine, norvaline,
ornithine, phenylalanine, proline,
pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine,
and valine. Amino acid
groups may be optionally substituted with one, two, three, or, in the case of
amino acid groups of two
carbons or more, four substituents independently selected from the group
consisting of: (1) C1_6
alkoxy; (2) C1_6 alkylsulfinyl; (3) amino, as defined herein (e.g.,
unsubstituted amino (i.e., -NH2) or a
substituted amino (i.e., -N(RN1)2, where RN1 is as defined for amino); (4)
C6_10 aryl-C1_6 alkoxy; (5)
azido; (6) halo; (7) (C2_9heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo
(e.g., carboxyaldehyde or
acyl); (11) C17 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) -CO2RA, where
RA is selected from the
group consisting of (a) C1_20 alkyl (e.g., C1_6 alkyl), (b) C2_20 alkenyl
(e.g., C2_6 alkenyl), (c) C6_10 aryl, (d)
hydrogen, (e) C1_6 alk-C6_10 aryl, (f) amino-C1_20 alkyl, (g) polyethylene
glycol of -
(C1-12)s2(OCH2C1-12)s1 (CI-12)s3OR', wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from 1 to 4),
each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to
4, from 0 to 6, from 1 to 4,
from 1 to 6, or from 1 to 10), and R' is H or C1_20 alkyl, and (h) amino-
polyethylene glycol of -
NRNi(C1-12)s2(CH2CH20)si(C1-12)s3NRN1, wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from
1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently,
hydrogen or optionally
substituted C1_6 alkyl; (15) -C(0)NRB Rc , where each of RB and IR is,
independently, selected from
the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl, and (d)
C1_6 alk-C6_10 aryl; (16) -
SO2RD, where RD is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) C1_6 alk-C6_10
aryl, and (d) hydroxy; (17) -SO2NRERF, where each of RE and RF is,
independently, selected from
the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl and (d)
C1_6 alk-C6_10 aryl; (18) -
C(0)RG , where RG is selected from the group consisting of (a) C1_20 alkyl
(e.g., C1_6 alkyl), (b) C2_20
alkenyl (e.g., C2_6 alkenyl), (c) C6_10 aryl, (d) hydrogen, (e) C1_6 alk-C6_10
aryl, (f) amino-C1_20 alkyl, (g)
polyethylene glycol of -(CH2)s2(OCH2C1-12)si(C1-12)s3OR', wherein s1 is an
integer from 1 to 10 (e.g.,
from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer
from 0 to 10 (e.g., from 0 to
4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R' is H or
C1_20 alkyl, and (h) amino-
polyethylene glycol of -NRN1 (C1-12)s2(CH2CH20)si (C1-12)s3NRN1, wherein s1 is
an integer from 1 to 10
- 193 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an
integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each
RN1 is, independently,
hydrogen or optionally substituted C1_6 alkyl; (19) -NRHC(0)RI', wherein RH'
is selected from the group
consisting of (al) hydrogen and (bl) C1_6 alkyl, and RI' is selected from the
group consisting of (a2)
C1_20 alkyl (e.g., C1_6 alkyl), (b2) C2_20 alkenyl (e.g., C2_6 alkenyl), (c2)
C6_10 aryl, (d2) hydrogen, (e2) C1_
6 alk-C6_10 aryl, (f2) amino-C1_20 alkyl, (g2) polyethylene glycol of -
(CH2)s2(OCH2CH2)s1(CH2)s3OR',
wherein sl is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each
of s2 and s3,
independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or
from 1 to 10), and R' is H or C1_20 alkyl, and (h2) amino-polyethylene glycol
of -
NRNi(CH2)s2(CH2CH20)si(C1-12)s3NRN1, wherein 51 is an integer from 1 to 10
(e.g., from 1 to 6 or from
1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g.,
from 0 to 4, from 0 to 6,
from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently,
hydrogen or optionally
substituted C1_6 alkyl; (20) -NRJ C(0)ORK', wherein RJ is selected from the
group consisting of (al)
hydrogen and (bl) C1_6 alkyl, and RK' is selected from the group consisting of
(a2) C1_20 alkyl (e.g., C1_6
alkyl), (b2) C2_20 alkenyl (e.g., C2_6 alkenyl), (c2) C6_10 aryl, (d2)
hydrogen, (e2) C1_6 alk-C6_10 aryl, (f2)
amino-C1_20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2C1-12)si(C1-
12)s3OR', wherein 51 is an
integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3,
independently, is an integer
from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or
from 1 to 10), and R' is H or C1_
alkyl, and (h2) amino-polyethylene glycol of -NRN1(CI-12)s2(CH2CH20)si(C1-
12)s3NRN1, wherein 51 is
20 an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2
and s3, independently, is an
integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to
6, or from 1 to 10), and each
-N1
h< is, independently, hydrogen or optionally substituted C1_6 alkyl; and
(21) amidine. In some
embodiments, each of these groups can be further substituted as described
herein.
The term "aminoalkoxy," as used herein, represents an alkoxy group, as defined
herein,
substituted by an amino group, as defined herein. The alkyl and amino each can
be further
substituted with 1, 2, 3, or 4 substituent groups as described herein for the
respective group (e.g.,
CO2RA', where RA' is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) hydrogen,
and (d) C1_6 alk-C6_10 aryl, e.g., carboxy).
The term "aminoalkyl," as used herein, represents an alkyl group, as defined
herein,
substituted by an amino group, as defined herein. The alkyl and amino each can
be further
substituted with 1, 2, 3, or 4 substituent groups as described herein for the
respective group (e.g.,
CO2RA', where RA' is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) hydrogen,
and (d) C1_6 alk-C6_10 aryl, e.g., carboxy, and/or an N-protecting group).
The term "aminoalkenyl," as used herein, represents an alkenyl group, as
defined herein,
substituted by an amino group, as defined herein. The alkenyl and amino each
can be further
substituted with 1, 2, 3, or 4 substituent groups as described herein for the
respective group (e.g.,
CO2RA', where RA' is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) hydrogen,
and (d) C1_6 alk-C6_10 aryl, e.g., carboxy, and/or an N-protecting group).
The term "aminoalkynyl," as used herein, represents an alkynyl group, as
defined herein,
substituted by an amino group, as defined herein. The alkynyl and amino each
can be further
substituted with 1, 2, 3, or 4 substituent groups as described herein for the
respective group (e.g.,
- 194 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
CO2RA', where RA' is selected from the group consisting of (a) C1_6 alkyl, (b)
C6_10 aryl, (c) hydrogen,
and (d) C1_6 alk-C6_10 aryl, e.g., carboxy, and/or an N-protecting group).
The term "aryl," as used herein, represents a mono-, bicyclic, or multicyclic
carbocyclic
ring system having one or two aromatic rings and is exemplified by phenyl,
naphthyl, 1,2-
dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, phenanthrenyl,
fluorenyl, indanyl, indenyl,
and the like, and may be optionally substituted with 1, 2, 3, 4, or 5
substituents independently
selected from the group consisting of: (1) C17 acyl (e.g., carboxyaldehyde);
(2) C1_20 alkyl (e.g., C1_6
alkyl, C1_6 alkoxy-C1_6 alkyl, C1_6 alkylsulfinyl-C1_6 alkyl, amino-C1_6
alkyl, azido-C1_6 alkyl,
(carboxyaldehyde)-C1_6 alkyl, halo-C1_6 alkyl (e.g., perfluoroalkyl), hydroxy-
C1_6 alkyl, nitro-C1_6 alkyl, or
C1_6 thioalkoxy-C1_6 alkyl); (3) C1_20 alkoxy (e.g., C1_6 alkoxy, such as
perfluoroalkoxy); (4) C1_6
alkylsulfinyl; (5) C6_10 aryl; (6) amino; (7) C1_6 alk-C6_10 aryl; (8) azido;
(9) C3_8 cycloalkyl; (10) C1_6 alk-
C3_8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., c1_12 heteroaryl);
(13) (Ci_12 heterocyclypoxY;
(14) hydroxy; (15) nitro; (16) C1_20 thioalkoxy (e.g., C1_6 thioalkoxy); (17)
¨(CH2)qCO2RA', where q is an
integer from zero to four, and RA' is selected from the group consisting of
(a) C1_6 alkyl, (b) C6_10 aryl,
(c) hydrogen, and (d) C1_6 alk-C6_10 aryl; (18) ¨(CH2)qCONRaRc', where q is an
integer from zero to
four and where R13 and Rc' are independently selected from the group
consisting of (a) hydrogen, (b)
C1_6 alkyl, (c) C6_10 aryl, and (d) C1_6 alk-C6_10 aryl; (19) ¨(C1-12)qS02RD',
where q is an integer from zero
to four and where R ' is selected from the group consisting of (a) alkyl, (b)
C6_10 aryl, and (c) alk-C6_10
aryl; (20) ¨(CH2)qS02NRE'RF', where q is an integer from zero to four and
where each of RE' and RF'
is, independently, selected from the group consisting of (a) hydrogen, (b)
C1_6 alkyl, (c) C6_10 aryl, and
(d) C1_6 alk-C6_10 aryl; (21) thiol; (22) C6_10 aryloxy; (23) c38 cycloalkoxy;
(24) C6_10 aryl-C1_6 alkoxy;
(25) C1_6 alk-C1_12 heterocyclyl (e.g., C1_6 alk-C1_12 heteroaryl); (26) C2_20
alkenyl; and (27) C2_20 alkynyl.
In some embodiments, each of these groups can be further substituted as
described herein. For
example, the alkylene group of a C1-alkaryl or a Cralkheterocyclylcan be
further substituted with an
oxo group to afford the respective aryloyl and (heterocyclypoyl substituent
group.
The term "arylalkoxy," as used herein, represents an alkaryl group, as defined
herein,
attached to the parent molecular group through an oxygen atom. Exemplary
unsubstituted arylalkoxy
groups include from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20
carbons, such as C6_10 aryl-C1_6
alkoxy, C6_10 aryl-C1_10 alkoxy, or c6-10 aryl-C1_20 alkoxy). In some
embodiments, the arylalkoxy group
can be substituted with 1, 2, 3, or 4 substituents as defined herein
The term "arylalkoxycarbonyl," as used herein, represents an arylalkoxy group,
as defined
herein, attached to the parent molecular group through a carbonyl (e.g., -C(0)-
0-alkyl-aryl).
Exemplary unsubstituted arylalkoxy groups include from 8 to 31 carbons (e.g.,
from 8 to 17 or from 8
to 21 carbons, such as C6_10 aryl-C1_6 alkoxy-carbonyl, C6_10 aryl-C1_10
alkoxy-carbonyl, or C6_10 aryl-C1-
20 alkoxy-carbonyl). In some embodiments, the arylalkoxycarbonyl group can be
substituted with 1, 2,
3, or 4 substituents as defined herein.
The term "aryloxy" represents a chemical substituent of formula ¨OR', where R'
is an aryl
group of 6 to 18 carbons, unless otherwise specified. In some embodiments, the
aryl group can be
substituted with 1, 2, 3, or 4 substituents as defined herein.
The term "aryloyl," as used herein, represents an aryl group, as defined
herein, that is
attached to the parent molecular group through a carbonyl group. Exemplary
unsubstituted aryloyl
- 195 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
groups are of 7 to 11 carbons. In some embodiments, the aryl group can be
substituted with 1, 2, 3,
or 4 substituents as defined herein.
The term "azido" represents an ¨N3 group, which can also be represented as
¨N=N=N.
The term "bicyclic," as used herein, refer to a structure having two rings,
which may be
aromatic or non-aromatic. Bicyclic structures include spirocyclyl groups, as
defined herein, and two
rings that share one or more bridges, where such bridges can include one atom
or a chain including
two, three, or more atoms. Exemplary bicyclic groups include a bicyclic
carbocyclyl group, where the
first and second rings are carbocyclyl groups, as defined herein; a bicyclic
aryl groups, where the first
and second rings are aryl groups, as defined herein; bicyclic heterocyclyl
groups, where the first ring
is a heterocyclyl group and the second ring is a carbocyclyl (e.g., aryl) or
heterocyclyl (e.g.,
heteroaryl) group; and bicyclic heteroaryl groups, where the first ring is a
heteroaryl group and the
second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl)
group. In some
embodiments, the bicyclic group can be substituted with 1, 2, 3, or 4
substituents as defined herein
for cycloalkyl, heterocyclyl, and aryl groups.
The term "boranyl," as used herein, represents ¨B(RB1)3, where each RB1 is,
independently, selected from the group consisting of H and optionally
substituted alkyl. In some
embodiments, the boranyl group can be substituted with 1, 2, 3, or 4
substituents as defined herein
for alkyl.
The terms "carbocyclic" and "carbocyclyl," as used herein, refer to an
optionally
substituted C3_12 monocyclic, bicyclic, or tricyclic structure in which the
rings, which may be aromatic
or non-aromatic, are formed by carbon atoms. Carbocyclic structures include
cycloalkyl, cycloalkenyl,
and aryl groups.
The term "carbamoyl," as used herein, represents ¨C(0)-N(RN1)2, where the
meaning of
each RN1 is found in the definition of "amino" provided herein.
The term "carbamoylalkyl," as used herein, represents an alkyl group, as
defined herein,
substituted by a carbamoyl group, as defined herein. The alkyl group can be
further substituted with
1, 2, 3, or 4 substituent groups as described herein.
The term "carbamyl," as used herein, refers to a carbamate group having the
structure
-NRN1C(=0)OR or -0C(=0)N(RN1)2, where the meaning of each RN1 is found in the
definition of
"amino" provided herein, and R is alkyl, cycloalkyl, alkcycloalkyl, aryl,
alkaryl, heterocyclyl (e.g.,
heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), as defined herein.
The term "carbonyl," as used herein, represents a C(0) group, which can also
be
represented as C=0.
The term "carboxyaldehyde" represents an acyl group having the structure ¨CHO.
The term "carboxy," as used herein, means ¨CO2H.
The term "carboxyalkoxy," as used herein, represents an alkoxy group, as
defined herein,
substituted by a carboxy group, as defined herein. The alkoxy group can be
further substituted with
1, 2, 3, or 4 substituent groups as described herein for the alkyl group, and
the carboxy group can be
optionally substituted with one or more 0-protecting groups.
The term "carboxyalkyl," as used herein, represents an alkyl group, as defined
herein,
substituted by a carboxy group, as defined herein. The alkyl group can be
further substituted with 1,
- 196 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
2, 3, or 4 substituent groups as described herein, and the carboxy group can
be optionally substituted
with one or more 0-protecting groups.
The term "carboxyaminoalkyl," as used herein, represents an aminoalkyl group,
as defined
herein, substituted by a carboxy, as defined herein. The carboxy, alkyl, and
amino each can be
further substituted with 1, 2, 3, or 4 substituent groups as described herein
for the respective group
(e.g., CO2RA', where RA' is selected from the group consisting of (a) C1_6
alkyl, (b) C6_10 aryl, (c)
hydrogen, and (d) C1_6 alk-C6_10 aryl, e.g., carboxy, and/or an N-protecting
group, and/or an 0-
protecting group).
The term "cyano," as used herein, represents an ¨CN group.
The term "cycloalkoxy" represents a chemical substituent of formula ¨OR, where
R is a C3_
8 cycloalkyl group, as defined herein, unless otherwise specified. The
cycloalkyl group can be further
substituted with 1, 2, 3, or 4 substituent groups as described herein.
Exemplary unsubstituted
cycloalkoxy groups are from 3 to 8 carbons. In some embodiment, the cycloalkyl
group can be
further substituted with 1, 2, 3, or 4 substituent groups as described herein.
The term "cycloalkyl," as used herein represents a monovalent saturated or
unsaturated
non-aromatic cyclic hydrocarbon group from three to eight carbons, unless
otherwise specified, and is
exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl,
bicycle heptyl, and the
like. When the cycloalkyl group includes one carbon-carbon double bond, the
cycloalkyl group can
be referred to as a "cycloalkenyl" group. Exemplary cycloalkenyl groups
include cyclopentenyl,
cyclohexenyl, and the like. The cycloalkyl groups of this invention can be
optionally substituted with:
(1) C1_7 acyl (e.g., carboxyaldehyde); (2) C1_20 alkyl (e.g., C1_6 alkyl, C1_6
alkoxy-C1_6 alkyl, C1_6
alkylsulfinyl-C1_6 alkyl, amino-C1_6 alkyl, azido-C1_6 alkyl,
(carboxyaldehyde)-C1_6 alkyl, halo-C1_6 alkyl
(e.g., perfluoroalkyl), hydroxy-C1_6 alkyl, nitro-C1_6 alkyl, or
C1_6thioalkoxy-C1_6 alkyl); (3) C1_20 alkoxy
(e.g., C1_6 alkoxy, such as perfluoroalkoxy); (4) C1_5 alkylsulfinyl; (5)
C6_10 aryl; (6) amino; (7) C1_6 alk-
C6_10 aryl; (8) azido; (9) C3_8 cycloalkyl; (10) C1_6 alk-C3_8 cycloalkyl;
(11) halo; (12) Ci_12 heterocyclyl
(e.g., C1_12 heteroaryl); (13) (C1_12 heterocyclypoxy; (14) hydroxy; (15)
nitro; (16) C1_20 thioalkoxy (e.g.,
C1_6 thioalkoxy); (17) ¨(CH2)qCO2RA', where q is an integer from zero to four,
and RA' is selected from
the group consisting of (a) C1_6 alkyl, (b) C6_10 aryl, (c) hydrogen, and (d)
C1_6 alk-C6_10 aryl; (18) ¨
"
(CH2)qC0NRBRc, where q is an integer from zero to four and where RB' and Ra
are independently
selected from the group consisting of (a) hydrogen, (b) C6_10 alkyl, (c) C6_10
aryl, and (d) C1_6 alk-C6_10
aryl; (19) ¨(CH2)qS02RD', where q is an integer from zero to four and where R
' is selected from the
group consisting of (a) C6_10 alkyl, (b) C6_10 aryl, and (c) C1_6 alk-C6_10
aryl; (20) ¨(CH2)qS02NRERF,
where q is an integer from zero to four and where each of RE' and RF' is,
independently, selected from
the group consisting of (a) hydrogen, (b) C6_10 alkyl, (c) C6_10 aryl, and (d)
C1_6 alk-C6_10 aryl; (21) thiol;
(22) C6_10 aryloxy; (23) C3_8 cycloalkoxy; (24) C6_10 aryl-C1_6 alkoxy; (25)
C1_6 alk-C1_12 heterocyclyl
(e.g., C1_6 alk-C1_12 heteroaryl); (26) oxo; (27) C2_20 alkenyl; and (28)
C2_20 alkynyl. In some
embodiments, each of these groups can be further substituted as described
herein. For example, the
alkylene group of a C1-alkaryl or a C1-alkheterocycly1 can be further
substituted with an oxo group to
afford the respective aryloyl and (heterocyclyl)oyl substituent group.
The term "diastereomer," as used herein means stereoisomers that are not
mirror images
of one another and are non-superimposable on one another.
- 197 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
The term "effective amount" of an agent, as used herein, is that amount
sufficient to effect
beneficial or desired results, for example, clinical results, and, as such, an
"effective amount"
depends upon the context in which it is being applied. For example, in the
context of administering
an agent that treats cancer, an effective amount of an agent is, for example,
an amount sufficient to
achieve treatment, as defined herein, of cancer, as compared to the response
obtained without
administration of the agent.
The term "enantiomer," as used herein, means each individual optically active
form of a
compound of the invention, having an optical purity or enantiomeric excess (as
determined by
methods standard in the art) of at least 80% (i.e., at least 90% of one
enantiomer and at most 10% of
the other enantiomer), preferably at least 90% and more preferably at least
98%.
The term "halo," as used herein, represents a halogen selected from bromine,
chlorine,
iodine, or fluorine.
The term "haloalkoxy," as used herein, represents an alkoxy group, as defined
herein,
substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkoxy may be
substituted with one, two,
three, or, in the case of alkyl groups of two carbons or more, four halogens.
Haloalkoxy groups
include perfluoroalkoxys (e.g., -0CF3), -OCHF2, -OCH2F, -OCCI3, -OCH2CH2Br, -
OCH2CH(CH2CH2Br)CH3, and -OCHICH3. In some embodiments, the haloalkoxy group
can be
further substituted with 1, 2, 3, or 4 substituent groups as described herein
for alkyl groups.
The term "haloalkyl," as used herein, represents an alkyl group, as defined
herein,
substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkyl may be
substituted with one, two,
three, or, in the case of alkyl groups of two carbons or more, four halogens.
Haloalkyl groups include
perfluoroalkyls (e.g., -CF3), -CHF2, -CH2F, -CCI3, -CH2CH2Br, -
CH2CH(CH2CH2BOCH3, and -CHICH3.
In some embodiments, the haloalkyl group can be further substituted with 1, 2,
3, or 4 substituent
groups as described herein for alkyl groups.
The term "heteroalkylene," as used herein, refers to an alkylene group, as
defined herein,
in which one or two of the constituent carbon atoms have each been replaced by
nitrogen, oxygen, or
sulfur. In some embodiments, the heteroalkylene group can be further
substituted with 1, 2, 3, or 4
substituent groups as described herein for alkylene groups.
The term "heteroaryl," as used herein, represents that subset of
heterocyclyls, as defined
herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the
mono- or multicyclic ring
system. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to
11, 1 to 10, 1 to 9, 2 to
12, 2 to 11, 2 to 10, or 2 to 9) carbons. In some embodiment, the heteroaryl
is substituted with 1, 2,
3, or 4 substituents groups as defined for a heterocyclyl group.
The term "heterocyclyl," as used herein represents a 5-, 6- or 7-membered
ring, unless
otherwise specified, containing one, two, three, or four heteroatoms
independently selected from the
group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero
to two double
bonds, and the 6- and 7-membered rings have zero to three double bonds.
Exemplary unsubstituted
heterocyclyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2
to 11, 2 to 10, or 2 to 9)
carbons. The term "heterocyclyl" also represents a heterocyclic compound
having a bridged
multicyclic structure in which one or more carbons and/or heteroatoms bridges
two non-adjacent
members of a monocyclic ring, e.g., a quinuclidinyl group. The term
"heterocyclyl" includes bicyclic,
- 198 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
tricyclic, and tetracyclic groups in which any of the above heterocyclic rings
is fused to one, two, or
three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene
ring, a cyclopentane ring,
a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl,
quinolyl, isoquinolyl,
tetrahydroquinolyl, benzofuryl, benzothienyl and the like. Examples of fused
heterocyclyls include
tropanes and 1,2,3,5,8,8a-hexahydroindolizine. Heterocyclics include pyrrolyl,
pyrrolinyl, pyrrolidinyl,
pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl,
imidazolidinyl, pyridyl, piperidinyl,
homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl,
oxazolidinyl, isoxazolyl,
isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl,
isothiazolyl, isothiazolidinyl,
indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, dihydroquinoxalinyl,
quinazolinyl, cinnolinyl,
phthalazinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzothiadiazolyl,
fury!, thienyl,
thiazolidinyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl (e.g., 1,2,3-
oxadiazoly1), purinyl, thiadiazolyl
(e.g., 1,2,3-thiadiazoly1), tetrahydrofuranyl, dihydrofuranyl,
tetrahydrothienyl, dihydrothienyl,
dihydroindolyl, dihydroquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl,
dihydroisoquinolyl, pyranyl,
dihydropyranyl, dithiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, and
the like, including
dihydro and tetrahydro forms thereof, where one or more double bonds are
reduced and replaced
with hydrogens. Still other exemplary heterocyclyls include: 2,3,4,5-
tetrahydro-2-oxo-oxazoly1; 2,3-
dihydro-2-oxo-1H-imidazoly1; 2,3,4,5-tetrahydro-5-oxo-1H-pyrazoly1 (e.g.,
2,3,4,5-tetrahydro-2-pheny1-
5-oxo-1H-pyrazoly1); 2,3,4,5-tetrahydro-2,4-dioxo-1H-imidazoly1 (e.g., 2,3,4,5-
tetrahydro-2,4-dioxo-5-
methy1-5-pheny1-1H-imidazoly1); 2,3-dihydro-2-thioxo-1,3,4-oxadiazoly1(e.g.,
2,3-dihydro-2-thioxo-5-
phenyl-1,3,4-oxadiazoly1); 4,5-dihydro-5-oxo-1H-triazoly1 (e.g., 4,5-dihydro-3-
methyl-4-amino 5-oxo-
1H-triazoly1); 1,2,3,4-tetrahydro-2,4-dioxopyridinyl (e.g., 1,2,3,4-tetrahydro-
2,4-dioxo-3,3-
diethylpyridinyl); 2,6-dioxo-piperidinyl (e.g., 2,6-dioxo-3-ethyl-3-
phenylpiperidinyl); 1,6-dihydro-6-
oxopyridiminyl; 1,6-dihydro-4-oxopyrimidinyl (e.g., 2-(methylthio)-1,6-dihydro-
4-oxo-5-
methylpyrimidin-1-y1); 1,2,3,4-tetrahydro-2,4-dioxopyrimidinyl (e.g., 1,2,3,4-
tetrahydro-2,4-dioxo-3-
ethylpyrimidinyl); 1,6-dihydro-6-oxo-pyridazinyl (e.g., 1,6-dihydro-6-oxo-3-
ethylpyridazinyl); 1,6-
dihydro-6-oxo-1,2,4-triazinyl (e.g., 1,6-dihydro-5-isopropy1-6-oxo-1,2,4-
triazinyl); 2,3-dihydro-2-oxo-
1H-indoly1 (e.g., 3,3-dimethy1-2,3-dihydro-2-oxo-1H-indoly1 and 2,3-dihydro-2-
oxo-3,3'-spiropropane-
1H-indo1-1-y1); 1,3-dihydro-1-oxo-2H-iso-indoly1; 1,3-dihydro-1,3-dioxo-2H-iso-
indoly1; 1H-
benzopyrazoly1(e.g., 1-(ethoxycarbony1)- 1H-benzopyrazoly1); 2,3-dihydro-2-oxo-
1H-benzimidazoly1
(e.g., 3-ethy1-2,3-dihydro-2-oxo-1H-benzimidazoly1); 2,3-dihydro-2-oxo-
benzoxazoly1 (e.g., 5-chloro-
2,3-dihydro-2-oxo-benzoxazoly1); 2,3-dihydro-2-oxo-benzoxazoly1; 2-oxo-2H-
benzopyranyl; 1,4-
benzodioxanyl; 1,3-benzodioxanyl; 2,3-dihydro-3-oxo,4H-1,3-benzothiazinyl; 3,4-
dihydro-4-oxo-3H-
quinazolinyl (e.g., 2-methyl-3,4-dihydro-4-oxo-3H-quinazolinyl); 1,2,3,4-
tetrahydro-2,4-dioxo-3H-
quinazolyl (e.g., 1-ethyl-1,2,3,4-tetrahydro-2,4-dioxo-3H-quinazoly1); 1,2,3,6-
tetrahydro-2,6-dioxo-7H-
purinyl (e.g., 1,2,3,6-tetrahydro-1,3-dimethy1-2,6-dioxo-7 H -purinyl);
1,2,3,6-tetrahydro-2,6-dioxo-1 H
¨purinyl (e.g., 1,2,3,6-tetrahydro-3,7-dimethy1-2,6-dioxo-1 H -purinyl); 2-
oxobenz[c,d]indoly1; 1,1-
dioxo-2H-naphth[1,8-c,d]isothiazoly1; and 1,8-naphthylenedicarboxamido.
Additional heterocyclics
include 3,3a,4,5,6,6a-hexahydro-pyrrolo[3,4-b]pyrrol-(2H)-yl, and 2,5-
diazabicyclo[2.2.1]heptan-2-yl,
homopiperazinyl (or diazepanyl), tetrahydropyranyl, dithiazolyl, benzofuranyl,
benzothienyl, oxepanyl,
thiepanyl, azocanyl, oxecanyl, and thiocanyl. Heterocyclic groups also include
groups of the formula
- 199 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Fx'
oG'
, where
E' is selected from the group consisting of -N- and -CH-; F' is selected from
the group
consisting of -N=CH-, -NH-CH2-, -NH-C(0)-, -NH-, -CH=N-, -CH2-NH-, -C(0)-NH-, -
CH=CH-, -CH2-, -
CH2CH2-, -CH20-, -OCH2-, -0-, and -S-; and G' is selected from the group
consisting of -CH- and -N-.
Any of the heterocyclyl groups mentioned herein may be optionally substituted
with one, two, three,
four or five substituents independently selected from the group consisting of:
(1) C17 acyl (e.g.,
carboxyaldehyde ); (2) C1_20 alkyl (e.g., C1_6 alkyl, Ci_6 alkoxy-C1_6 alkyl,
C1_6 alkylsulfinyl-C1_6 alkyl,
amino-C1_6 alkyl, azido-C1_6 alkyl, (carboxyaldehyde)-Ci_6 alkyl, halo-C1_6
alkyl (e.g., perfluoroalkyl),
hydroxy-C1_6 alkyl, nitro-C1_6 alkyl, or C1_6 thioalkoxy-C1_6 alkyl); (3)
C1_20 alkoxy (e.g., C1_6 alkoxy, such
as perfluoroalkoxy); (4) C1_6 alkylsulfinyl; (5) C6_10 aryl; (6) amino; (7)
C1_6 alk-C6_10 aryl; (8) azido; (9)
C3_8 cycloalkyl; (10) C1_6 alk-C3_8 cycloalkyl; (11) halo; (12) Ci_12
heterocyclyl (e.g., c2_12 heteroaryl);
(13) (C1_12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1_20 thioalkoxy
(e.g., C1_6thioalkoxy); (17)
-(CH2)qCO2RA , where q is an integer from zero to four, and RA is selected
from the group consisting
of (a) C1_6 alkyl, (b) C6_10 aryl, (c) hydrogen, and (d) C1_6 alk-C6_10 aryl;
(18) -(CH2)qCONRBRc, where q
is an integer from zero to four and where RB and Rc are independently selected
from the group
consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl, and (d) C1_6 alk-
C6_10 aryl; (19) -(CH2)cS02RD,
where q is an integer from zero to four and where RD is selected from the
group consisting of (a) C1_6
alkyl, (b) C6_10 aryl, and (c) C1_6 alk-C6_10 aryl; (20) (CH2)c,S02NRERF ,
where q is an integer from zero
to four and where each of RE and RF is, independently, selected from the group
consisting of (a)
hydrogen, (b) C1_6 alkyl, (c) C6_10 aryl, and (d) C1_6 alk-C6_10 aryl; (21)
thiol; (22) C6_10 aryloxy; (23) Cm
cycloalkoxy; (24) arylalkoxy; (25) C1_6 alk-C1_12 heterocyclyl (e.g., C1_6 alk-
C1_12 heteroaryl); (26) oxo;
(27) (Ci_12 heterocyclyl)imino; (28) C2_20 alkenyl; and (29) C2_20 alkynyl. In
some embodiments, each
of these groups can be further substituted as described herein. For example,
the alkylene group of a
C1-alkaryl or a C1-alkheterocycly1 can be further substituted with an oxo
group to afford the respective
aryloyl and (heterocyclyl)oyl substituent group.
The term "(heterocyclyl) imino," as used herein, represents a heterocyclyl
group, as
defined herein, attached to the parent molecular group through an imino group.
In some
embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4
substituent groups as
defined herein.
The term "(heterocyclyl)oxy," as used herein, represents a heterocyclyl group,
as defined
herein, attached to the parent molecular group through an oxygen atom. In some
embodiments, the
heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as
defined herein.
The term "(heterocyclyl)oyl," as used herein, represents a heterocyclyl group,
as defined
herein, attached to the parent molecular group through a carbonyl group. In
some embodiments, the
heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as
defined herein.
The term "hydrocarbon," as used herein, represents a group consisting only of
carbon and
hydrogen atoms.
- 200 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
The term "hydroxy," as used herein, represents an ¨OH group. In some
embodiments, the
hydroxy group can be substituted with 1, 2, 3, or 4 substituent groups (e.g.,
0-protecting groups) as
defined herein for an alkyl.
The term "hydroxyalkenyl," as used herein, represents an alkenyl group, as
defined
herein, substituted by one to three hydroxy groups, with the proviso that no
more than one hydroxy
group may be attached to a single carbon atom of the alkyl group, and is
exemplified by
dihydroxypropenyl, hydroxyisopentenyl, and the like. In some embodiments, the
hydroxyalkenyl
group can be substituted with 1, 2, 3, or 4 substituent groups (e.g., 0-
protecting groups) as defined
herein for an alkyl.
The term "hydroxyalkyl," as used herein, represents an alkyl group, as defined
herein,
substituted by one to three hydroxy groups, with the proviso that no more than
one hydroxy group
may be attached to a single carbon atom of the alkyl group, and is exemplified
by hydroxymethyl,
dihydroxypropyl, and the like. In some embodiments, the hydroxyalkyl group can
be substituted with
1, 2, 3, or 4 substituent groups (e.g., 0-protecting groups) as defined herein
for an alkyl.
The term "hydroxyalkynyl," as used herein, represents an alkynyl group, as
defined herein,
substituted by one to three hydroxy groups, with the proviso that no more than
one hydroxy group
may be attached to a single carbon atom of the alkyl group. In some
embodiments, the
hydroxyalkynyl group can be substituted with 1, 2, 3, or 4 substituent groups
(e.g., 0-protecting
groups) as defined herein for an alkyl.
The term "isomer," as used herein, means any tautomer, stereoisomer,
enantiomer, or
diastereomer of any compound of the invention. It is recognized that the
compounds of the invention
can have one or more chiral centers and/or double bonds and, therefore, exist
as stereoisomers,
such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers
(e.g., enantiomers (i.e.,
(+) or (-)) or cis/trans isomers). According to the invention, the chemical
structures depicted herein,
and therefore the compounds of the invention, encompass all of the
corresponding stereoisomers,
that is, both the stereomerically pure form (e.g., geometrically pure,
enantiomerically pure, or
diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g.,
racemates.
Enantiomeric and stereoisomeric mixtures of compounds of the invention can
typically be resolved
into their component enantiomers or stereoisomers by well-known methods, such
as chiral-phase gas
chromatography, chiral-phase high performance liquid chromatography,
crystallizing the compound
as a chiral salt complex, or crystallizing the compound in a chiral solvent.
Enantiomers and
stereoisomers can also be obtained from stereomerically or enantiomerically
pure intermediates,
reagents, and catalysts by well-known asymmetric synthetic methods.
The term "N-protected amino," as used herein, refers to an amino group, as
defined
herein, to which is attached one or two N-protecting groups, as defined
herein.
The term "N-protecting group," as used herein, represents those groups
intended to
protect an amino group against undesirable reactions during synthetic
procedures. Commonly used
N-protecting groups are disclosed in Greene, "Protective Groups in Organic
Synthesis," 3rd Edition
(John Wiley & Sons, New York, 1999), which is incorporated herein by
reference. N-protecting
groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl,
propionyl, pivaloyl, t-
butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl,
phthalyl, o-
- 201 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl,
4-nitrobenzoyl, and
chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids
such as alanine, leucine,
phenylalanine, and the like; sulfonyl-containing groups such as
benzenesulfonyl, p-toluenesulfonyl,
and the like; carbamate forming groups such as benzyloxycarbonyl, p-
chlorobenzyloxycarbonyl,
p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-
nitrobenzyloxycarbonyl,
p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-
dimethoxybenzyloxycarbonyl, 2,4-
dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-
dimethoxybenzyloxycarbonyl,
3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyI)-1-methylethoxycarbonyl,
a,a-dimethy1-
3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl,
diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl,
methoxycarbonyl,
allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-
nitrophenoxy carbonyl, fluoreny1-
9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl,
cyclohexyloxycarbonyl,
phenylthiocarbonyl, and the like, alkaryl groups such as benzyl,
triphenylmethyl, benzyloxymethyl,
and the like and silyl groups, such as trimethylsilyl, and the like. Preferred
N-protecting groups are
formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl,
benzyl, t-butyloxycarbonyl (Boc),
and benzyloxycarbonyl (Cbz).
The term "nitro," as used herein, represents an ¨NO2 group.
The term "0-protecting group," as used herein, represents those groups
intended to
protect an oxygen containing (e.g., phenol, hydroxyl, or carbonyl) group
against undesirable reactions
during synthetic procedures. Commonly used 0-protecting groups are disclosed
in Greene,
"Protective Groups in Organic Synthesis," 3rd Edition (John Wiley & Sons, New
York, 1999), which is
incorporated herein by reference. Exemplary 0-protecting groups include acyl,
aryloyl, or carbamyl
groups, such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-
chloroacetyl, 2-bromoacetyl,
trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-
chlorobutyryl, benzoyl, 4-
chlorobenzoyl, 4-bromobenzoyl, t-butyldimethylsilyl, tri-iso-
propylsilyloxymethyl, 4,4'-dimethoxytrityl,
isobutyryl, phenoxyacetyl, 4-isopropylpehenoxyacetyl, dimethylformamidino, and
4-nitrobenzoyl;
alkylcarbonyl groups, such as acyl, acetyl, propionyl, pivaloyl, and the like;
optionally substituted
arylcarbonyl groups, such as benzoyl; silyl groups, such as trimethylsilyl
(TMS), tert-butyldimethylsilyl
(TBDMS), tri-iso-propylsilyloxymethyl (TOM), triisopropylsilyl (TIPS), and the
like; ether-forming
groups with the hydroxyl, such methyl, methoxymethyl, tetrahydropyranyl,
benzyl, p-methoxybenzyl,
trityl, and the like; alkoxycarbonyls, such as methoxycarbonyl,
ethoxycarbonyl, isopropoxycarbonyl, n-
isopropoxycarbonyl, n-butyloxycarbonyl, isobutyloxycarbonyl, sec-
butyloxycarbonyl, t-
butyloxycarbonyl, 2-ethylhexyloxycarbonyl, cyclohexyloxycarbonyl,
methyloxycarbonyl, and the like;
alkoxyalkoxycarbonyl groups, such as methoxymethoxycarbonyl,
ethoxymethoxycarbonyl, 2-
methoxyethoxycarbonyl, 2-ethoxyethoxycarbonyl, 2-butoxyethoxycarbonyl, 2-
methoxyethoxymethoxycarbonyl, allyloxycarbonyl, propargyloxycarbonyl, 2-
butenoxycarbonyl, 3-
methy1-2-butenoxycarbonyl, and the like; haloalkoxycarbonyls, such as 2-
chloroethoxycarbonyl, 2-
chloroethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, and the like; optionally
substituted
arylalkoxycarbonyl groups, such as benzyloxycarbonyl, p-
methylbenzyloxycarbonyl, p-
methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2,4-
dinitrobenzyloxycarbonyl, 3,5-
dimethylbenzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-bromobenzyloxy-
carbonyl,
- 202 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
fluorenylmethyloxycarbonyl, and the like; and optionally substituted
aryloxycarbonyl groups, such as
phenoxycarbonyl, p-nitrophenoxycarbonyl, o-nitrophenoxycarbonyl, 2,4-
dinitrophenoxycarbonyl, p-
methyl-phenoxycarbonyl, m-methylphenoxycarbonyl, o-bromophenoxycarbonyl, 3,5-
dimethylphenoxycarbonyl, p-chlorophenoxycarbonyl, 2-chloro-4-nitrophenoxy-
carbonyl, and the like);
substituted alkyl, aryl, and alkaryl ethers (e.g., trityl; methylthiomethyl;
methoxymethyl;
benzyloxymethyl; siloxymethyl; 2,2,2,-trichloroethoxymethyl;
tetrahydropyranyl; tetrahydrofuranyl;
ethoxyethyl; 142-(trimethylsilypethoxy]ethyl; 2-trimethylsilylethyl; t-butyl
ether; p-chlorophenyl, p-
methoxyphenyl, p-nitrophenyl, benzyl, p-methoxybenzyl, and nitrobenzyl); silyl
ethers (e.g.,
trimethylsilyl; triethylsilyl; triisopropylsilyl; dimethylisopropylsilyl; t-
butyldimethylsilyl; t-
butyldiphenylsilyl; tribenzylsilyl; triphenylsilyl; and diphenymethylsilyl);
carbonates (e.g., methyl,
methoxymethyl, 9-fluorenylmethyl; ethyl; 2,2,2-trichloroethyl; 2-
(trimethylsilyl)ethyl; vinyl, ally!,
nitrophenyl; benzyl; methoxybenzyl; 3,4-dimethoxybenzyl; and nitrobenzyl);
carbonyl-protecting
groups (e.g., acetal and ketal groups, such as dimethyl acetal, 1,3-dioxolane,
and the like; acylal
groups; and dithiane groups, such as 1,3-dithianes, 1,3-dithiolane, and the
like); carboxylic acid-
protecting groups (e.g., ester groups, such as methyl ester, benzyl ester, t-
butyl ester, orthoesters,
and the like; and oxazoline groups.
The term "oxo" as used herein, represents =0.
The term "perfluoroalkyl," as used herein, represents an alkyl group, as
defined herein,
where each hydrogen radical bound to the alkyl group has been replaced by a
fluoride radical.
Perfluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl,
and the like.
The term "perfluoroalkoxy," as used herein, represents an alkoxy group, as
defined herein,
where each hydrogen radical bound to the alkoxy group has been replaced by a
fluoride radical.
Perfluoroalkoxy groups are exemplified by trifluoromethoxy, pentafluoroethoxy,
and the like.
The term "spirocyclyl," as used herein, represents a C27 alkylene diradical,
both ends of
which are bonded to the same carbon atom of the parent group to form a
spirocyclic group, and also
a C1_6 heteroalkylene diradical, both ends of which are bonded to the same
atom. The heteroalkylene
radical forming the spirocyclyl group can containing one, two, three, or four
heteroatoms
independently selected from the group consisting of nitrogen, oxygen, and
sulfur. In some
embodiments, the spirocyclyl group includes one to seven carbons, excluding
the carbon atom to
which the diradical is attached. The spirocyclyl groups of the invention may
be optionally substituted
with 1, 2, 3, or 4 substituents provided herein as optional substituents for
cycloalkyl and/or
heterocyclyl groups.
The term "stereoisomer," as used herein, refers to all possible different
isomeric as well as
conformational forms which a compound may possess (e.g., a compound of any
formula described
herein), in particular all possible stereochemically and conformationally
isomeric forms, all
diastereomers, enantiomers and/or conformers of the basic molecular structure.
Some compounds of
the present invention may exist in different tautomeric forms, all of the
latter being included within the
scope of the present invention.
The term "sulfoalkyl," as used herein, represents an alkyl group, as defined
herein,
substituted by a sulfo group of ¨503H. In some embodiments, the alkyl group
can be further
- 203 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
substituted with 1, 2, 3, or 4 substituent groups as described herein, and the
sulfo group can be
further substituted with one or more 0-protecting groups (e.g., as described
herein).
The term "sulfonyl," as used herein, represents an -S(0)2- group.
The term "thioalkaryl," as used herein, represents a chemical substituent of
formula ¨SR,
where R is an alkaryl group. In some embodiments, the alkaryl group can be
further substituted with
1, 2, 3, or 4 substituent groups as described herein.
The term "thioalkheterocyclyl," as used herein, represents a chemical
substituent of
formula ¨SR, where R is an alkheterocyclyl group. In some embodiments, the
alkheterocyclyl group
can be further substituted with 1, 2, 3, or 4 substituent groups as described
herein.
The term "thioalkoxy," as used herein, represents a chemical substituent of
formula ¨SR,
where R is an alkyl group, as defined herein. In some embodiments, the alkyl
group can be further
substituted with 1, 2, 3, or 4 substituent groups as described herein.
Compound: As used herein, the term "compound," is meant to include all
stereoisomers,
geometric isomers, tautomers, and isotopes of the structures depicted.
The compounds described herein can be asymmetric (e.g., having one or more
stereocenters). All stereoisomers, such as enantiomers and diastereomers, are
intended unless
otherwise indicated. Compounds of the present disclosure that contain
asymmetrically substituted
carbon atoms can be isolated in optically active or racemic forms. Methods on
how to prepare
optically active forms from optically active starting materials are known in
the art, such as by
resolution of racemic mixtures or by stereoselective synthesis. Many geometric
isomers of olefins,
C=N double bonds, and the like can also be present in the compounds described
herein, and all such
stable isomers are contemplated in the present disclosure. Cis and trans
geometric isomers of the
compounds of the present disclosure are described and may be isolated as a
mixture of isomers or
as separated isomeric forms.
Compounds of the present disclosure also include tautomeric forms. Tautomeric
forms
result from the swapping of a single bond with an adjacent double bond and the
concomitant
migration of a proton. Tautomeric forms include prototropic tautomers which
are isomeric protonation
states having the same empirical formula and total charge. Examples
prototropic tautomers include
ketone ¨ enol pairs, amide ¨ imidic acid pairs, lactam ¨ lactim pairs, amide ¨
imidic acid pairs,
enamine ¨ imine pairs, and annular forms where a proton can occupy two or more
positions of a
heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-
triazole, 1H- and 2H-
isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or
sterically locked into
one form by appropriate substitution.
Compounds of the present disclosure also include all of the isotopes of the
atoms
occurring in the intermediate or final compounds. "Isotopes" refers to atoms
having the same atomic
number but different mass numbers resulting from a different number of
neutrons in the nuclei. For
example, isotopes of hydrogen include tritium and deuterium.
The compounds and salts of the present disclosure can be prepared in
combination with
solvent or water molecules to form solvates and hydrates by routine methods.
Conserved: As used herein, the term "conserved" refers to nucleotides or amino
acid
residues of a polynucleotide sequence or polypeptide sequence, respectively,
that are those that
- 204 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
occur unaltered in the same position of two or more sequences being compared.
Nucleotides or
amino acids that are relatively conserved are those that are conserved amongst
more related
sequences than nucleotides or amino acids appearing elsewhere in the
sequences.
In some embodiments, two or more sequences are said to be "completely
conserved" if
they are 100% identical to one another. In some embodiments, two or more
sequences are said to
be "highly conserved" if they are at least 70% identical, at least 80%
identical, at least 90% identical,
or at least 95% identical to one another. In some embodiments, two or more
sequences are said to
be "highly conserved" if they are about 70% identical, about 80% identical,
about 90% identical, about
95%, about 98%, or about 99% identical to one another. In some embodiments,
two or more
sequences are said to be "conserved" if they are at least 30% identical, at
least 40% identical, at least
50% identical, at least 60% identical, at least 70% identical, at least 80%
identical, at least 90%
identical, or at least 95% identical to one another. In some embodiments, two
or more sequences are
said to be "conserved" if they are about 30% identical, about 40% identical,
about 50% identical,
about 60% identical, about 70% identical, about 80% identical, about 90%
identical, about 95%
identical, about 98% identical, or about 99% identical to one another.
Conservation of sequence may
apply to the entire length of an oligonucleotide or polypeptide or may apply
to a portion, region or
feature thereof.
Cyclic or Cyclized: As used herein, the term "cyclic" refers to the presence
of a continuous
loop. Cyclic molecules need not be circular, only joined to form an unbroken
chain of subunits. Cyclic
molecules such as the mRNA of the present invention may be single units or
multimers or comprise
one or more components of a complex or higher order structure.
Cytostatic: As used herein, "cytostatic" refers to inhibiting, reducing,
suppressing the
growth, division, or multiplication of a cell (e.g., a mammalian cell (e.g., a
human cell)), bacterium,
virus, fungus, protozoan, parasite, prion, or a combination thereof.
Cytotoxic: As used herein, "cytotoxic" refers to killing or causing injurious,
toxic, or deadly
effect on a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium,
virus, fungus, protozoan,
parasite, prion, or a combination thereof.
Delivery: As used herein, "delivery" refers to the act or manner of delivering
a compound,
substance, entity, moiety, cargo or payload.
Delivery Agent: As used herein, "delivery agent" refers to any substance which
facilitates,
at least in part, the in vivo delivery of a polynucleotide to targeted cells.
Destabilized: As used herein, the term "destable," "destabilize," or
"destabilizing region"
means a region or molecule that is less stable than a starting, wild-type or
native form of the same
region or molecule.
Detectable label: As used herein, "detectable label" refers to one or more
markers,
signals, or moieties which are attached, incorporated or associated with
another entity that is readily
detected by methods known in the art including radiography, fluorescence,
chemiluminescence,
enzymatic activity, absorbance and the like. Detectable labels include
radioisotopes, fluorophores,
chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin,
streptavidin and haptens,
quantum dots, and the like. Detectable labels may be located at any position
in the peptides or
- 205 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
proteins disclosed herein. They may be within the amino acids, the peptides,
or proteins, or located
at the N- or C- termini.
Digest: As used herein, the term "digest" means to break apart into smaller
pieces or
components. When referring to polypeptides or proteins, digestion results in
the production of
peptides.
Distal: As used herein, the term "distal" means situated away from the center
or away from
a point or region of interest.
Encoded protein cleavage signal: As used herein, "encoded protein cleavage
signal"
refers to the nucleotide sequence which encodes a protein cleavage signal.
Engineered: As used herein, embodiments of the invention are "engineered" when
they
are designed to have a feature or property, whether structural or chemical,
that varies from a starting
point, wild type or native molecule.
Expression: As used herein, "expression" of a nucleic acid sequence refers to
one or
more of the following events: (1) production of an RNA template from a DNA
sequence (e.g., by
transcription); (2) processing of an RNA transcript (e.g., by splicing,
editing, 5' cap formation, and/or
3' end processing); (3) translation of an RNA into a polypeptide or protein;
and (4) post-translational
modification of a polypeptide or protein.
Feature: As used herein, a "feature" refers to a characteristic, a property,
or a distinctive
element.
Formulation: As used herein, a "formulation" includes at least a
polynucleotide and a
delivery agent.
Fragment: A "fragment," as used herein, refers to a portion. For example,
fragments of
proteins may comprise polypeptides obtained by digesting full-length protein
isolated from cultured
cells.
Functional: As used herein, a "functional" biological molecule is a biological
molecule in a
form in which it exhibits a property and/or activity by which it is
characterized.
Homology: As used herein, the term "homology" refers to the overall
relatedness between
polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules
and/or RNA
molecules) and/or between polypeptide molecules. In some embodiments,
polymeric molecules are
considered to be "homologous" to one another if their sequences are at least
25%, 30%, 35%, 40%,
45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or
similar. The term
"homologous" necessarily refers to a comparison between at least two sequences
(polynucleotide or
polypeptide sequences). In accordance with the invention, two polynucleotide
sequences are
considered to be homologous if the polypeptides they encode are at least about
50%, 60%, 70%,
80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino
acids. In some
embodiments, homologous polynucleotide sequences are characterized by the
ability to encode a
stretch of at least 4-5 uniquely specified amino acids. For polynucleotide
sequences less than 60
nucleotides in length, homology is determined by the ability to encode a
stretch of at least 4-5
uniquely specified amino acids. In accordance with the invention, two protein
sequences are
considered to be homologous if the proteins are at least about 50%, 60%, 70%,
80%, or 90%
identical for at least one stretch of at least about 20 amino acids.
- 206 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Identity: As used herein, the term "identity" refers to the overall
relatedness between
polymeric molecules, e.g., between oligonucleotide molecules (e.g. DNA
molecules and/or RNA
molecules) and/or between polypeptide molecules. Calculation of the percent
identity of two
polynucleotide sequences, for example, can be performed by aligning the two
sequences for optimal
comparison purposes (e.g., gaps can be introduced in one or both of a first
and a second nucleic acid
sequences for optimal alignment and non-identical sequences can be disregarded
for comparison
purposes). In certain embodiments, the length of a sequence aligned for
comparison purposes is at
least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least
80%, at least 90%, at least
95%, or 100% of the length of the reference sequence. The nucleotides at
corresponding nucleotide
positions are then compared. When a position in the first sequence is occupied
by the same
nucleotide as the corresponding position in the second sequence, then the
molecules are identical at
that position. The percent identity between the two sequences is a function of
the number of identical
positions shared by the sequences, taking into account the number of gaps, and
the length of each
gap, which needs to be introduced for optimal alignment of the two sequences.
The comparison of
sequences and determination of percent identity between two sequences can be
accomplished using
a mathematical algorithm. For example, the percent identity between two
nucleotide sequences can
be determined using methods such as those described in Computational Molecular
Biology, Lesk, A.
M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics
and Genome Projects,
Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in
Molecular Biology, von
Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I,
Griffin, A. M., and
Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis
Primer, Gribskov, M.
and Devereux, J., eds., M Stockton Press, New York, 1991. For example, the
percent identity
between two nucleotide sequences can be determined using the algorithm of
Meyers and Miller
(CABIOS, 1989, 4:11-17), which has been incorporated into the ALIGN program
(version 2.0) using a
PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of
4. The percent identity
between two nucleotide sequences can, alternatively, be determined using the
GAP program in the
GCG software package using an NWSgapdna.CMP matrix. Methods commonly employed
to
determine percent identity between sequences include, but are not limited to
those disclosed in
Carillo, H., and Lipman, D., SIAM J Applied Math., 48:1073 (1988);
incorporated herein by reference.
Techniques for determining identity are codified in publicly available
computer programs. Exemplary
computer software to determine homology between two sequences include, but are
not limited to,
GCG program package, Devereux, J., etal., Nucleic Acids Research, 12(1), 387
(1984)), BLASTP,
BLASTN, and FASTA Altschul, S. F. etal., J. Molec. Biol., 215, 403 (1990)).
Inhibit expression of a gene: As used herein, the phrase "inhibit expression
of a gene"
means to cause a reduction in the amount of an expression product of the gene.
The expression
product can be an RNA transcribed from the gene (e.g., an mRNA) or a
polypeptide translated from
an mRNA transcribed from the gene. Typically a reduction in the level of an
mRNA results in a
reduction in the level of a polypeptide translated therefrom. The level of
expression may be
determined using standard techniques for measuring mRNA or protein.
- 207 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
In vitro: As used herein, the term "in vitro" refers to events that occur in
an artificial
environment, e.g., in a test tube or reaction vessel, in cell culture, in a
Petri dish, etc., rather than
within an organism (e.g., animal, plant, or microbe).
In vivo: As used herein, the term "in vivo" refers to events that occur within
an organism
(e.g., animal, plant, or microbe or cell or tissue thereof).
Isolated: As used herein, the term "isolated" refers to a substance or entity
that has been
separated from at least some of the components with which it was associated
(whether in nature or in
an experimental setting). Isolated substances may have varying levels of
purity in reference to the
substances from which they have been associated. Isolated substances and/or
entities may be
separated from at least about 10%, about 20%, about 30%, about 40%, about 50%,
about 60%,
about 70%, about 80%, about 90%, or more of the other components with which
they were initially
associated. In some embodiments, isolated agents are more than about 80%,
about 85%, about
90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about
97%, about 98%,
about 99%, or more than about 99% pure. As used herein, a substance is "pure"
if it is substantially
free of other components. Substantially isolated: By "substantially isolated"
is meant that the
compound is substantially separated from the environment in which it was
formed or detected. Partial
separation can include, for example, a composition enriched in the compound of
the present
disclosure. Substantial separation can include compositions containing at
least about 50%, at least
about 60%, at least about 70%, at least about 80%, at least about 90%, at
least about 95%, at least
about 97%, or at least about 99% by weight of the compound of the present
disclosure, or salt
thereof. Methods for isolating compounds and their salts are routine in the
art.
Linker: As used herein, a linker refers to a group of atoms, e.g., 10-1,000
atoms, and can
be comprised of the atoms or groups such as, but not limited to, carbon,
amino, alkylamino, oxygen,
sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached
to a modified nucleoside or
nucleotide on the nucleobase or sugar moiety at a first end, and to a payload,
e.g., a detectable or
therapeutic agent, at a second end. The linker may be of sufficient length as
to not interfere with
incorporation into a nucleic acid sequence. The linker can be used for any
useful purpose, such as to
form multimers (e.g., through linkage of two or more polynucleotides) or
conjugates, as well as to
administer a payload, as described herein. Examples of chemical groups that
can be incorporated
into the linker include, but are not limited to, alkyl, alkenyl, alkynyl,
amido, amino, ether, thioether,
ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be
optionally substituted, as
described herein. Examples of linkers include, but are not limited to,
unsaturated alkanes,
polyethylene glycols (e.g., ethylene or propylene glycol monomeric units,
e.g., diethylene glycol,
dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene
glycol, or tetraethylene glycol),
and dextran polymers, Other examples include, but are not limited to,
cleavable moieties within the
linker, such as, for example, a disulfide bond (-S-S-) or an azo bond (-N=N-),
which can be cleaved
using a reducing agent or photolysis. Non-limiting examples of a selectively
cleavable bond include
an amido bond can be cleaved for example by the use of tris(2-
carboxyethyl)phosphine (TCEP), or
other reducing agents, and/or photolysis, as well as an ester bond can be
cleaved for example by
acidic or basic hydrolysis.
- 208 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Modified: As used herein "modified" refers to a changed state or structure of
a molecule of
the invention. Molecules may be modified in many ways including chemically,
structurally, and
functionally. In one embodiment, the mRNA molecules of the present invention
are modified by the
introduction of non-natural nucleosides and/or nucleotides, e.g., as it
relates to the natural
ribonucleotides A, U, G, and C. Noncanonical nucleotides such as the cap
structures are not
considered "modified" although they differ from the chemical structure of the
A, C, G, U
ribonucleotides.
Naturally occurring: As used herein, "naturally occurring" means existing in
nature without
artificial aid.
Non-human vertebrate: As used herein, a "non human vertebrate" includes all
vertebrates
except Homo sapiens, including wild and domesticated species. Examples of non-
human vertebrates
include, but are not limited to, mammals, such as alpaca, banteng, bison,
camel, cat, cattle, deer,
dog, donkey, gayal, goat, guinea pig, horse, llama, mule, pig, rabbit,
reindeer, sheep water buffalo,
and yak.
Off-target: As used herein, "off target" refers to any unintended effect on
any one or more
target, gene, or cellular transcript.
Open reading frame: As used herein, "open reading frame" or "ORF" refers to a
sequence
which does not contain a stop codon in a given reading frame.
Operably linked: As used herein, the phrase "operably linked" refers to a
functional
connection between two or more molecules, constructs, transcripts, entities,
moieties or the like.
Paratope: As used herein, a "paratope" refers to the antigen-binding site of
an antibody.
Patient: As used herein, "patient" refers to a subject who may seek or be in
need of
treatment, requires treatment, is receiving treatment, will receive treatment,
or a subject who is under
care by a trained professional for a particular disease or condition.
Optionally substituted: Herein a phrase of the form "optionally substituted X"
(e.g.,
optionally substituted alkyl) is intended to be equivalent to "X, wherein X is
optionally substituted"
(e.g., "alkyl, wherein said alkyl is optionally substituted"). It is not
intended to mean that the feature
"X" (e.g. alkyl) per se is optional.
Peptide: As used herein, "peptide" is less than or equal to 50 amino acids
long, e.g., about
5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
Pharmaceutically acceptable: The phrase "pharmaceutically acceptable" is
employed
herein to refer to those compounds, materials, compositions, and/or dosage
forms which are, within
the scope of sound medical judgment, suitable for use in contact with the
tissues of human beings
and animals without excessive toxicity, irritation, allergic response, or
other problem or complication,
commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable excipients: The phrase "pharmaceutically
acceptable
excipient," as used herein, refers any ingredient other than the compounds
described herein (for
example, a vehicle capable of suspending or dissolving the active compound)
and having the
properties of being substantially nontoxic and non-inflammatory in a patient.
Excipients may include,
for example: antiadherents, antioxidants, binders, coatings, compression aids,
disintegrants, dyes
(colors), emollients, emulsifiers, fillers (diluents), film formers or
coatings, flavors, fragrances, glidants
- 209 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(flow enhancers), lubricants, preservatives, printing inks, sorbents,
suspensing or dispersing agents,
sweeteners, and waters of hydration. Exemplary excipients include, but are not
limited to: butylated
hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium
stearate,
croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone,
cysteine, ethylcellulose,
gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose,
magnesium stearate,
maltitol, mannitol, methionine, methylcellulose, methyl paraben,
microcrystalline cellulose,
polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch,
propyl paraben, retinyl
palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium
citrate, sodium starch
glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium
dioxide, vitamin A, vitamin E,
vitamin C, and xylitol.
Pharmaceutically acceptable salts: The present disclosure also includes
pharmaceutically
acceptable salts of the compounds described herein. As used herein,
"pharmaceutically acceptable
salts" refers to derivatives of the disclosed compounds wherein the parent
compound is modified by
converting an existing acid or base moiety to its salt form (e.g., by reacting
the free base group with a
suitable organic acid). Examples of pharmaceutically acceptable salts include,
but are not limited to,
mineral or organic acid salts of basic residues such as amines; alkali or
organic salts of acidic
residues such as carboxylic acids; and the like. Representative acid addition
salts include acetate,
adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate,
bisulfate, borate, butyrate,
camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate,
dodecylsulfate,
ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate,
heptonate, hexanoate,
hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate,
lactobionate, lactate, laurate,
lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-
naphthalenesulfonate, nicotinate,
nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-
phenylpropionate, phosphate,
picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate,
thiocyanate, toluenesulfonate,
undecanoate, valerate salts, and the like. Representative alkali or alkaline
earth metal salts include
sodium, lithium, potassium, calcium, magnesium, and the like, as well as
nontoxic ammonium,
quaternary ammonium, and amine cations, including, but not limited to
ammonium,
tetramethylammonium, tetraethylammonium, methylamine, dimethylamine,
trimethylamine,
triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts
of the present
disclosure include the conventional non-toxic salts of the parent compound
formed, for example, from
non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of
the present disclosure
can be synthesized from the parent compound which contains a basic or acidic
moiety by
conventional chemical methods. Generally, such salts can be prepared by
reacting the free acid or
base forms of these compounds with a stoichiometric amount of the appropriate
base or acid in water
or in an organic solvent, or in a mixture of the two; generally, nonaqueous
media like ether, ethyl
acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of
suitable salts are found in
Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company,
Easton, Pa., 1985, p.
1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and
C.G. Wermuth (eds.),
Wiley-VCH, 2008, and Berge et al., Journal of Pharmaceutical Science, 66, 1-19
(1977).
Pharmacokinetic: As used herein, "pharmacokinetic" refers to any one or more
properties
of a molecule or compound as it relates to the determination of the fate of
substances administered to
- 210 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
a living organism. Pharmacokinetics is divided into several areas including
the extent and rate of
absorption, distribution, metabolism and excretion. This is commonly referred
to as ADME where: (A)
Absorption is the process of a substance entering the blood circulation; (D)
Distribution is the
dispersion or dissemination of substances throughout the fluids and tissues of
the body; (M)
Metabolism (or Biotransformation) is the irreversible transformation of parent
compounds into
daughter metabolites; and (E) Excretion (or Elimination) refers to the
elimination of the substances
from the body. In rare cases, some drugs irreversibly accumulate in body
tissue.
Pharmaceutically acceptable solvate: The term "pharmaceutically acceptable
solvate," as
used herein, means a compound of the invention wherein molecules of a suitable
solvent are
incorporated in the crystal lattice. A suitable solvent is physiologically
tolerable at the dosage
administered. For example, solvates may be prepared by crystallization,
recrystallization, or
precipitation from a solution that includes organic solvents, water, or a
mixture thereof. Examples of
suitable solvents are ethanol, water (for example, mono-, di-, and tri-
hydrates), N-methylpyrrolidinone
(NMP), dimethyl sulfoxide (DMSO), N,N'-dimethylformamide (DMF), N,N'-
dimethylacetamide
(DMAC), 1,3-dimethy1-2-imidazolidinone (DMEU), 1,3-dimethy1-3,4,5,6-tetrahydro-
2-(1H)-
pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate,
benzyl alcohol, 2-
pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the
solvate is referred to as a
"hydrate."
Physicochemical: As used herein, "physicochemical" means of or relating to a
physical
and/or chemical property.
Preventing: As used herein, the term "preventing" refers to partially or
completely delaying
onset of an infection, disease, disorder and/or condition; partially or
completely delaying onset of one
or more symptoms, features, or clinical manifestations of a particular
infection, disease, disorder,
and/or condition; partially or completely delaying onset of one or more
symptoms, features, or
manifestations of a particular infection, disease, disorder, and/or condition;
partially or completely
delaying progression from an infection, a particular disease, disorder and/or
condition; and/or
decreasing the risk of developing pathology associated with the infection, the
disease, disorder,
and/or condition.
Prodrug: The present disclosure also includes prodrugs of the compounds
described
herein. As used herein, "prodrugs" refer to any substance, molecule or entity
which is in a form
predicate for that substance, molecule or entity to act as a therapeutic upon
chemical or physical
alteration. Prodrugs may by covalently bonded or sequestered in some way and
which release or are
converted into the active drug moiety prior to, upon or after administered to
a mammalian subject.
Prodrugs can be prepared by modifying functional groups present in the
compounds in such a way
that the modifications are cleaved, either in routine manipulation or in vivo,
to the parent compounds.
Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl
groups are bonded to
any group that, when administered to a mammalian subject, cleaves to form a
free hydroxyl, amino,
sulfhydryl, or carboxyl group respectively. Preparation and use of prodrugs is
discussed in T. Higuchi
and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S.
Symposium Series, and in
Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American
Pharmaceutical Association
and Pergamon Press, 1987.
- 211 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Proliferate: As used herein, the term "proliferate" means to grow, expand or
increase or
cause to grow, expand or increase rapidly. "Proliferative" means having the
ability to proliferate. "Anti-
proliferative" means having properties counter to or inapposite to
proliferative properties.
Protein cleavage site: As used herein, "protein cleavage site" refers to a
site where
controlled cleavage of the amino acid chain can be accomplished by chemical,
enzymatic or
photochemical means.
Protein cleavage signal: As used herein "protein cleavage signal" refers to at
least one
amino acid that flags or marks a polypeptide for cleavage.
Protein of interest: As used herein, the terms "proteins of interest" or
"desired proteins"
include those provided herein and fragments, mutants, variants, and
alterations thereof.
Proximal: As used herein, the term "proximal" means situated nearer to the
center or to a
point or region of interest.
Purified: As used herein, "purify," "purified," "purification" means to make
substantially pure
or clear from unwanted components, material defilement, admixture or
imperfection.
Sample: As used herein, the term "sample" or "biological sample" refers to a
subset of its
tissues, cells or component parts (e.g. body fluids, including but not limited
to blood, mucus,
lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid,
amniotic cord blood, urine,
vaginal fluid and semen). A sample further may include a homogenate, lysate or
extract prepared
from a whole organism or a subset of its tissues, cells or component parts, or
a fraction or portion
thereof, including but not limited to, for example, plasma, serum, spinal
fluid, lymph fluid, the external
sections of the skin, respiratory, intestinal, and genitourinary tracts,
tears, saliva, milk, blood cells,
tumors, organs. A sample further refers to a medium, such as a nutrient broth
or gel, which may
contain cellular components, such as proteins or nucleic acid molecule.
Signal Sequences: As used herein, the phrase "signal sequences" refers to a
sequence
which can direct the transport or localization of a protein.
Significant or Significantly: As used herein, the terms "significant" or
"significantly" are
used synonymously with the term "substantially."
Single unit dose: As used herein, a "single unit dose" is a dose of any
therapeutic
administed in one dose/at one time/single route/single point of contact, i.e.,
single administration
event.
Similarity: As used herein, the term "similarity" refers to the overall
relatedness between
polymeric molecules, e.g. between polynucleotide molecules (e.g. DNA molecules
and/or RNA
molecules) and/or between polypeptide molecules. Calculation of percent
similarity of polymeric
molecules to one another can be performed in the same manner as a calculation
of percent identity,
except that calculation of percent similarity takes into account conservative
substitutions as is
understood in the art.
Split dose: As used herein, a "split dose" is the division of single unit dose
or total daily
dose into two or more doses.
Stable: As used herein "stable" refers to a compound that is sufficiently
robust to survive
isolation to a useful degree of purity from a reaction mixture, and preferably
capable of formulation
into an efficacious therapeutic agent.
- 212 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Stabilized: As used herein, the term "stabilize", "stabilized," "stabilized
region" means to
make or become stable.
Subject: As used herein, the term "subject" or "patient" refers to any
organism to which a
composition in accordance with the invention may be administered, e.g., for
experimental, diagnostic,
prophylactic, and/or therapeutic purposes. Typical subjects include animals
(e.g., mammals such as
mice, rats, rabbits, non-human primates, and humans) and/or plants.
Substantially: As used herein, the term "substantially" refers to the
qualitative condition of
exhibiting total or near-total extent or degree of a characteristic or
property of interest. One of
ordinary skill in the biological arts will understand that biological and
chemical phenomena rarely, if
ever, go to completion and/or proceed to completeness or achieve or avoid an
absolute result. The
term "substantially" is therefore used herein to capture the potential lack of
completeness inherent in
many biological and chemical phenomena.
Substantially equal: As used herein as it relates to time differences between
doses, the
term means plus/minus 2%.
Substantially simultaneously: As used herein and as it relates to plurality of
doses, the
term means within 2 seconds.
Suffering from: An individual who is "suffering from" a disease, disorder,
and/or condition
has been diagnosed with or displays one or more symptoms of a disease,
disorder, and/or condition.
Susceptible to: An individual who is "susceptible to" a disease, disorder,
and/or condition
has not been diagnosed with and/or may not exhibit symptoms of the disease,
disorder, and/or
condition but harbors a propensity to develop a disease or its symptoms. In
some embodiments, an
individual who is susceptible to a disease, disorder, and/or condition (for
example, cancer) may be
characterized by one or more of the following: (1) a genetic mutation
associated with development of
the disease, disorder, and/or condition; (2) a genetic polymorphism associated
with development of
the disease, disorder, and/or condition; (3) increased and/or decreased
expression and/or activity of a
protein and/or nucleic acid associated with the disease, disorder, and/or
condition; (4) habits and/or
lifestyles associated with development of the disease, disorder, and/or
condition; (5) a family history
of the disease, disorder, and/or condition; and (6) exposure to and/or
infection with a microbe
associated with development of the disease, disorder, and/or condition. In
some embodiments, an
individual who is susceptible to a disease, disorder, and/or condition will
develop the disease,
disorder, and/or condition. In some embodiments, an individual who is
susceptible to a disease,
disorder, and/or condition will not develop the disease, disorder, and/or
condition.
Synthetic: The term "synthetic" means produced, prepared, and/or manufactured
by the
hand of man. Synthesis of polynucleotides or polypeptides or other molecules
of the present invention
may be chemical or enzymatic.
Targeted Cells: As used herein, "targeted cells" refers to any one or more
cells of interest.
The cells may be found in vitro, in vivo, in situ or in the tissue or organ of
an organism. The organism
may be an animal, preferably a mammal, more preferably a human and most
preferably a patient.
Therapeutic Agent: The term "therapeutic agent" refers to any agent that, when
administered to a subject, has a therapeutic, diagnostic, and/or prophylactic
effect and/or elicits a
desired biological and/or pharmacological effect.
- 213 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Therapeutically effective amount: As used herein, the term "therapeutically
effective
amount" means an amount of an agent to be delivered (e.g., nucleic acid, drug,
therapeutic agent,
diagnostic agent, prophylactic agent, etc.) that is sufficient, when
administered to a subject suffering
from or susceptible to an infection, disease, disorder, and/or condition, to
treat, improve symptoms of,
diagnose, prevent, and/or delay the onset of the infection, disease, disorder,
and/or condition.
Therapeutically effective outcome: As used herein, the term "therapeutically
effective
outcome" means an outcome that is sufficient in a subject suffering from or
susceptible to an
infection, disease, disorder, and/or condition, to treat, improve symptoms of,
diagnose, prevent,
and/or delay the onset of the infection, disease, disorder, and/or condition.
Total daily dose: As used herein, a "total daily dose" is an amount given or
prescribed in
24 hr period. It may be administered as a single unit dose.
Transcription factor: As used herein, the term "transcription factor" refers
to a DNA-binding
protein that regulates transcription of DNA into RNA, for example, by
activation or repression of
transcription. Some transcription factors effect regulation of transcription
alone, while others act in
concert with other proteins. Some transcription factor can both activate and
repress transcription
under certain conditions. In general, transcription factors bind a specific
target sequence or
sequences highly similar to a specific consensus sequence in a regulatory
region of a target gene.
Transcription factors may regulate transcription of a target gene alone or in
a complex with other
molecules.
Treating: As used herein, the term "treating" refers to partially or
completely alleviating,
ameliorating, improving, relieving, delaying onset of, inhibiting progression
of, reducing severity of,
and/or reducing incidence of one or more symptoms or features of a particular
infection, disease,
disorder, and/or condition. For example, "treating" cancer may refer to
inhibiting survival, growth,
and/or spread of a tumor. Treatment may be administered to a subject who does
not exhibit signs of
a disease, disorder, and/or condition and/or to a subject who exhibits only
early signs of a disease,
disorder, and/or condition for the purpose of decreasing the risk of
developing pathology associated
with the disease, disorder, and/or condition.
Unmodified: As used herein, "unmodified" refers to any substance, compound or
molecule
prior to being changed in any way. Unmodified may, but does not always, refer
to the wild type or
native form of a biomolecule. Molecules may undergo a series of modifications
whereby each
modified molecule may serve as the "unmodified" starting molecule for a
subsequent modification.
Equivalents and Scope
Those skilled in the art will recognize, or be able to ascertain using no more
than routine
experimentation, many equivalents to the specific embodiments in accordance
with the invention
described herein. The scope of the present invention is not intended to be
limited to the above
Description, but rather is as set forth in the appended claims.
In the claims, articles such as "a," "an," and "the" may mean one or more than
one unless
indicated to the contrary or otherwise evident from the context. Claims or
descriptions that include
"or" between one or more members of a group are considered satisfied if one,
more than one, or all of
the group members are present in, employed in, or otherwise relevant to a
given product or process
- 214 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
unless indicated to the contrary or otherwise evident from the context. The
invention includes
embodiments in which exactly one member of the group is present in, employed
in, or otherwise
relevant to a given product or process. The invention includes embodiments in
which more than one,
or all of the group members are present in, employed in, or otherwise relevant
to a given product or
process.
It is also noted that the term "comprising" is intended to be open and permits
but does not
require the inclusion of additional elements or steps. When the term
"comprising" is used herein, the
term "consisting of" is thus also encompassed and disclosed.
Where ranges are given, endpoints are included. Furthermore, it is to be
understood that
unless otherwise indicated or otherwise evident from the context and
understanding of one of
ordinary skill in the art, values that are expressed as ranges can assume any
specific value or
subrange within the stated ranges in different embodiments of the invention,
to the tenth of the unit of
the lower limit of the range, unless the context clearly dictates otherwise.
In addition, it is to be understood that any particular embodiment of the
present invention
that falls within the prior art may be explicitly excluded from any one or
more of the claims. Since
such embodiments are deemed to be known to one of ordinary skill in the art,
they may be excluded
even if the exclusion is not set forth explicitly herein. Any particular
embodiment of the compositions
of the invention (e.g., any nucleic acid or protein encoded thereby; any
method of production; any
method of use; etc.) can be excluded from any one or more claims, for any
reason, whether or not
related to the existence of prior art.
All cited sources, for example, references, publications, databases, database
entries, and
art cited herein, are incorporated into this application by reference, even if
not expressly stated in the
citation. In case of conflicting statements of a cited source and the instant
application, the statement
in the instant application shall control.
EXAMPLES
The present disclosure is further described in the following examples, which
do not limit
the scope of the disclosure described in the claims.
Example 1: PCR for cDNA Production
PCR procedures for the preparation of cDNA are performed using 2x KAPA HIFI TM
HotStart ReadyMix by Kapa Biosystems (Woburn, MA). This system includes 2x
KAPA
ReadyMix12.5 pl; Forward Primer (10 uM) 0.75 pl; Reverse Primer (10 uM) 0.75
pl; Template cDNA
100 ng; and dH20 diluted to 25.0 pl. The reaction conditions are at 95 C for
5 min. and 25 cycles of
98 C for 20 sec, then 58 C for 15 sec, then 72 C for 45 sec, then 72 C for
5 min. then 4 C to
termination.
The reverse primer of the instant invention incorporates a poly-T120 for a
poly-A120 in the
mRNA. Other reverse primers with longer or shorter poly-T tracts can be used
to adjust the length of
the poly-A tail in the mRNA.
The reaction is cleaned up using Invitrogen's PURELINKTM PCR Micro Kit
(Carlsbad, CA)
per manufacturer's instructions (up to 5 pg). Larger reactions will require a
cleanup using a product
- 215 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
with a larger capacity. Following the cleanup, the cDNA is quantified using
the NanoDrop and
analyzed by agarose gel electrophoresis to confirm the cDNA is the expected
size. The cDNA is then
submitted for sequencing analysis before proceeding to the in vitro
transcription reaction.
Example 2. In vitro Transcription (IVT)
A. Materials and Methods
Modified mRNAs according to the invention are made using standard laboratory
methods
and materials for in vitro transcription with the exception that the
nucleotide mix contains modified
nucleotides. The open reading frame (ORF) of the gene of interest may be
flanked by a 5'
untranslated region (UTR) containing a strong Kozak translational initiation
signal and an alpha-globin
3 UTR terminating with an oligo(dT) sequence for templated addition of a polyA
tail for mRNAs not
incorporating adenosine analogs. Adenosine-containing mRNAs are synthesized
without an oligo
(dT) sequence to allow for post-transcription poly (A) polymerase poly-(A)
tailing.
The ORF may also include various upstream or downstream additions (such as,
but not
limited to, 13-globin, tags, etc.) may be ordered from an optimization service
such as, but limited to,
DNA2.0 (Menlo Park, CA) and may contain multiple cloning sites which may have
Xbal recognition.
Upon receipt of the construct, it may be reconstituted and transformed into
chemically competent E.
coli.
For the present invention, NEB DH5-alpha Competent E. coli may be used.
Transformations are performed according to NEB instructions using 100 ng of
plasmid. The protocol
is as follows:
Thaw a tube of NEB 5-alpha Competent E. coli cells on ice for 10 minutes.
Add 1-5 pl containing 1 pg-100 ng of plasmid DNA to the cell mixture.
Carefully flick the
tube 4-5 times to mix cells and DNA. Do not vortex.
Place the mixture on ice for 30 minutes. Do not mix.
Heat shock at 42 C for exactly 30 seconds. Do not mix.
Place on ice for 5 minutes. Do not mix.
Pipette 950 pl of room temperature SOC into the mixture.
Place at 37 C for 60 minutes. Shake vigorously (250 rpm) or rotate.
Warm selection plates to 37 C.
Mix the cells thoroughly by flicking the tube and inverting.
Spread 50-100 pl of each dilution onto a selection plate and incubate
overnight at 37 C.
Alternatively, incubate at 30 C for 24-36 hours or 25 C for 48 hours.
A single colony is then used to inoculate 5 ml of LB growth media using the
appropriate
antibiotic and then allowed to grow (250 RPM, 37 C) for 5 hours. This is then
used to inoculate a 200
ml culture medium and allowed to grow overnight under the same conditions.
To isolate the plasmid (up to 850 pg), a maxi prep is performed using the
Invitrogen
PURELINKTM HiPure Maxiprep Kit (Carlsbad, CA), following the manufacturer's
instructions.
In order to generate cDNA for In Vitro Transcription (IVT), the plasmid is
first linearized
using a restriction enzyme such as Xbal. A typical restriction digest with
Xbal will comprise the
following: Plasmid 1.0 pg; 10x Buffer 1.0 pl; Xbal 1.5 pl; dH20 up to 10 pl;
incubated at 37 C for 1 hr.
- 216 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
If performing at lab scale (< 5pg), the reaction is cleaned up using
Invitrogen's PURELINKTM PCR
Micro Kit (Carlsbad, CA) per manufacturer's instructions. Larger scale
purifications may need to be
done with a product that has a larger load capacity such as Invitrogen's
standard PURELINKTM PCR
Kit (Carlsbad, CA). Following the cleanup, the linearized vector is quantified
using the NanoDrop and
analyzed to confirm linearization using agarose gel electrophoresis.
IVT Reaction
The in vitro transcription reaction generates mRNA containing modified
nucleotides or
modified RNA. The input nucleotide triphosphate (NTP) mix is made in-house
using natural and un-
natural NTPs.
A typical in vitro transcription reaction includes the following:
Template cDNA 1.0 pg
10x transcription buffer (400 mM Tris-HCI pH 8.0, 2.0 pl
190 mM MgC12, 50 mM DTT, 10 mM Spermidine)
Custom NTPs (25mM each 7.2 pl
RNase Inhibitor 20 U
T7 RNA polymerase 3000 U
dH20 up to 20.0 pl
Incubation at 37 C for 3 hr-5 hrs.
The crude IVT mix may be stored at 4 C overnight for cleanup the next day. 1
U of
RNase-free DNase is then used to digest the original template. After 15
minutes of incubation at 37
C, the mRNA is purified using Ambion's MEGACLEARTM Kit (Austin, TX) following
the manufacturer's
instructions. This kit can purify up to 500 pg of RNA. Following the cleanup,
the RNA is quantified
using the NanoDrop and analyzed by agarose gel electrophoresis to confirm the
RNA is the proper
size and that no degradation of the RNA has occurred.
The T7 RNA polymerase may be selected from, T7 RNA polymerase, T3 RNA
polymerase
and mutant polymerases such as, but not limited to, the novel polymerases able
to incorporate
modified NTPs as well as those polymerases described by Liu (Esvelt etal.
(Nature (2011)
472(7344):499-503 and U.S. Publication No. 20110177495) which recognize
alternate promoters,
Ellington (Chelliserrykattil and Ellington, Nature Biotechnology (2004)
22(9):1155-1160) describing a
T7 RNA polymerase variant to transcribe 2'-0-methyl RNA and Sousa (Padilla and
Sousa, Nucleic
Acids Research (2002) 30(24): e128) describing a T7 RNA polymerase double
mutant; herein
incorporated by reference in their entireties.
B. Agarose Gel Electrophoresis of modified mRNA
Individual modified mRNAs (200-400 ng in a 20 pl volume) are loaded into a
well on a
non-denaturing 1.2% Agarose E-Gel (Invitrogen, Carlsbad, CA) and run for 12-15
minutes according
to the manufacturer protocol.
- 217 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
C. Agarose Gel Electrophoresis of RT-PCR products
Individual reverse transcribed-PCR products (200-400ng) are loaded into a well
of a non-
denaturing 1.2% Agarose E-Gel (Invitrogen, Carlsbad, CA) and run for 12-15
minutes according to
the manufacturer protocol.
D. Nanodrop modified mRNA quantification and UV spectral data
Modified mRNAs in TE buffer (1 pl) are used for Nanodrop UV absorbance
readings to
quantitate the yield of each modified mRNA from an in vitro transcription
reaction (UV absorbance
traces are not shown).
Example 3. Enzymatic Capping of mRNA
Capping of the mRNA is performed as follows where the mixture includes: IVT
RNA 60 pg-
180pg and dH20 up to 72 pl. The mixture is incubated at 65 C for 5 minutes to
denature RNA, and
then is transferred immediately to ice.
The protocol then involves the mixing of 10x Capping Buffer (0.5 M Tris-HCI
(pH 8.0), 60
mM KCI, 12.5 mM MgC12) (10.0 pl); 20 mM GTP (5.0 pl); 20 mM S-Adenosyl
Methionine (2.5 pl);
RNase Inhibitor (100 U); 2'-0-Methyltransferase (400U); Vaccinia capping
enzyme (Guanylyl
transferase) (40 U); dH20 (Up to 28 pl); and incubation at 37 C for 30
minutes for 60 pg RNA or up to
2 hours for 180 pg of RNA.
The mRNA is then purified using Ambion's MEGACLEARTM Kit (Austin, TX)
following the
manufacturer's instructions. Following the cleanup, the RNA is quantified
using the NANODROPTM
(ThermoFisher, Waltham, MA) and analyzed by agarose gel electrophoresis to
confirm the RNA is the
proper size and that no degradation of the RNA has occurred. The RNA product
may also be
sequenced by running a reverse-transcription-PCR to generate the cDNA for
sequencing.
Example 4. 5'-Guanosine Capping
A. Materials and Methods
The cloning, gene synthesis and vector sequencing may be performed by DNA2.0
Inc.
(Menlo Park, CA). The ORF is restriction digested using Xbal and used for cDNA
synthesis using
tailed-or tail-less-PCR. The tailed-PCR cDNA product is used as the template
for the modified mRNA
synthesis reaction using 25mM each modified nucleotide mix (all modified
nucleotides may be custom
synthesized or purchased from TriLink Biotech, San Diego, CA except pyrrolo-C
triphosphate which
may be purchased from Glen Research, Sterling VA; unmodifed nucleotides are
purchased from
Epicenter Biotechnologies, Madison, WI) and CellScript MEGASCRIPTTm (Epicenter
Biotechnologies,
Madison, WI) complete mRNA synthesis kit.
The in vitro transcription reaction is run for 4 hours at 37 C. Modified mRNAs
incorporating adenosine analogs are poly (A) tailed using yeast Poly (A)
Polymerase (Affymetrix,
Santa Clara, CA). The PCR reaction uses HiFi PCR 2X MASTER MIXTM (Kapa
Biosystems, Woburn,
MA). Modified mRNAs are post-transcriptionally capped using recombinant
Vaccinia Virus Capping
Enzyme (New England BioLabs, Ipswich, MA) and a recombinant 2'-0-
methyltransferase (Epicenter
Biotechnologies, Madison, WI) to generate the 5'-guanosine Cap1 structure. Cap
2 structure and
- 218 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Cap 2 structures may be generated using additional 2'-0-methyltransferases.
The In vitro transcribed
mRNA product is run on an agarose gel and visualized. Modified mRNA may be
purified with
Ambion/Applied Biosystems (Austin, TX) MEGAClear RNATM purification kit. The
PCR uses
PURELINKTM PCR purification kit (Invitrogen, Carlsbad, CA). The product is
quantified on
NANODROPTM UV Absorbance (ThermoFisher, Waltham, MA). Quality, UV absorbance
quality and
visualization of the product was performed on an 1.2% agarose gel. The product
is resuspended in
TE buffer.
B. 5' Capping Modified Nucleic Acid (mRNA) Structure
5'-capping of modified mRNA may be completed concomitantly during the in vitro-
transcription reaction using the following chemical RNA cap analogs to
generate the 5'-guanosine cap
structure according to manufacturer protocols: 3"-0-Me-m7G(5)ppp(5')G (the
ARCA cap);
G(5')ppp(5')A; G(5')ppp(5')G; m7G(5')ppp(5')A; m7G(5')ppp(5')G (New England
BioLabs, Ipswich,
MA). 5'-capping of modified mRNA may be completed post-transcriptionally using
a Vaccinia Virus
Capping Enzyme to generate the "Cap 0" structure: m7G(5')ppp(5')G (New England
BioLabs, Ipswich,
MA). Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme
and a 2'-0
methyl-transferase to generate: m7G(5)ppp(5')G-2'-0-methyl. Cap 2 structure
may be generated
from the Cap 1 structure followed by the 2'-o-methylation of the 5'-
antepenultimate nucleotide using a
2'-0 methyl-transferase. Cap 3 structure may be generated from the Cap 2
structure followed by the
2'-o-methylation of the 5'-preantepenultimate nucleotide using a 2'-0 methyl-
transferase. Enzymes
are preferably derived from a recombinant source.
When transfected into mammalian cells, the modified mRNAs have a stability of
12-18
hours or more than 18 hours, e.g., 24, 36, 48, 60, 72 or greater than 72
hours.
Example 5. PolyA Tailing Reaction
Without a poly-T in the cDNA, a poly-A tailing reaction must be performed
before cleaning
the final product. This is done by mixing Capped IVT RNA (100 pl); RNase
Inhibitor (20 U); 10x
Tailing Buffer (0.5 M Tris-HCI (pH 8.0), 2.5 M NaCI, 100 mM MgC12)(12.0 pl);
20 mM ATP (6.0 pl);
Poly-A Polymerase (20 U); dH20 up to 123.5 pl and incubation at 37 C for 30
min. If the poly-A tail is
already in the transcript, then the tailing reaction may be skipped and
proceed directly to cleanup with
Ambion's MEGACLEARTM kit (Austin, TX) (up to 500 pg). Poly-A Polymerase is
preferably a
recombinant enzyme expressed in yeast.
For studies performed and described herein, the poly-A tail is encoded in the
IVT template
to comprise160 nucleotides in length. However, it should be understood that
the processivity or
integrity of the poly-A tailing reaction may not always result in exactly 160
nucleotides. Hence poly-A
tails of approximately 160 nucleotides, acid about 150-165, 155, 156, 157,
158, 159, 160, 161, 162,
163, 164 or 165 are within the scope of the invention.
- 219 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 6. Method of Screening for Protein Expression
A. Electrospray Ionization
A biological sample which may contain proteins encoded by modified RNA
administered to
the subject is prepared and analyzed according to the manufacturer protocol
for electrospray
ionization (ESI) using 1, 2, 3 or 4 mass analyzers. A biologic sample may also
be analyzed using a
tandem ESI mass spectrometry system.
Patterns of protein fragments, or whole proteins, are compared to known
controls for a
given protein and identity is determined by comparison.
B. Matrix-Assisted Laser Desorption/lonization
A biological sample which may contain proteins encoded by modified RNA
administered
to the subject is prepared and analyzed according to the manufacturer protocol
for matrix-assisted
laser desorption/ionization (MALDI).
Patterns of protein fragments, or whole proteins, are compared to known
controls for a
given protein and identity is determined by comparison.
C. Liquid Chromatography-Mass spectrometry-Mass spectrometry
A biological sample, which may contain proteins encoded by modified RNA, may
be
treated with a trypsin enzyme to digest the proteins contained within. The
resulting peptides are
analyzed by liquid chromatography-mass spectrometry-mass spectrometry
(LC/MS/MS). The
peptides are fragmented in the mass spectrometer to yield diagnostic patterns
that can be matched to
protein sequence databases via computer algorithms. The digested sample may be
diluted to achieve
1 ng or less starting material for a given protein. Biological samples
containing a simple buffer
background (e.g. water or volatile salts) are amenable to direct in-solution
digest; more complex
backgrounds (e.g. detergent, non-volatile salts, glycerol) require an
additional clean-up step to
facilitate the sample analysis.
Patterns of protein fragments, or whole proteins, are compared to known
controls for a
given protein and identity is determined by comparison.
Example 7. Transfection
A. Reverse Trans fection
For experiments performed in a 24-well collagen-coated tissue culture plate,
Keratinocytes
or other cells are seeded at a cell density of 1 x 105. For experiments
performed in a 96-well
collagen-coated tissue culture plate, Keratinocytes are seeded at a cell
density of 0.5 x 105. For each
modified mRNA to be transfected, modified mRNA: RNAIMAXTm are prepared as
described and
mixed with the cells in the multi-well plate within 6 hours of cell seeding
before cells had adhered to
the tissue culture plate.
- 220 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
B. Forward Transfection
In a 24-well collagen-coated tissue culture plate, Cells are seeded at a cell
density of 0.7
x 105. For experiments performed in a 96-well collagen-coated tissue culture
plate, Keratinocytes, if
used, are seeded at a cell density of 0.3 x 105. Cells are then grown to a
confluency of >70% for over
24 hours. For each modified mRNA to be transfected, modified mRNA: RNAIMAXTm
are prepared as
described and transfected onto the cells in the multi-well plate over 24 hours
after cell seeding and
adherence to the tissue culture plate.
C. Translation Screen: ELISA
Cells are grown in EpiLife medium with Supplement S7 from Invitrogen at a
confluence of
>70%. Cells are reverse transfected with 300 ng of the indicated chemically
modified mRNA
complexed with RNAIMAXTm from Invitrogen. Alternatively, cells are forward
transfected with 300 ng
modified mRNA complexed with RNAIMAXTm from Invitrogen. The RNA: RNAIMAXTm
complex is
formed by first incubating the RNA with Supplement-free EPILIFE media in a 5X
volumetric dilution
for 10 minutes at room temperature.
In a second vial, RNAIMAXTm reagent is incubated with Supplement-free EPILIFE
Media
in a 10X volumetric dilution for 10 minutes at room temperature. The RNA vial
is then mixed with the
RNAIMAXTm vial and incubated for 20-30 at room temperature before being added
to the cells in a
drop-wise fashion. Secreted polypeptide concentration in the culture medium is
measured at 18
hours post-transfection for each of the chemically modified mRNAs in
triplicate. Secretion of the
polypeptide of interest from transfected human cells is quantified using an
ELISA kit from Invitrogen
or R&D Systems (Minneapolis, MN) following the manufacturers recommended
instructions.
D. Dose and Duration: ELISA
Cells are grown in EPILIFE medium with Supplement S7 from Invitrogen at a
confluence
of >70%. Cells are reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng,
375ng, 750ng, or
150Ong modified mRNA complexed with RNAIMAXTm from Invitrogen. The modified
mRNA:
RNAIMAXTm complex is formed as described. Secreted polypeptide concentration
in the culture
medium is measured at 0, 6, 12, 24, and 48 hours post-transfection for each
concentration of each
modified mRNA in triplicate. Secretion of the polypeptide of interest from
transfected human cells is
quantified using an ELISA kit from Invitrogen or R&D Systems following the
manufacturers
recommended instructions.
Example 8. Cellular Innate Immune Response: IFN-beta ELISA and TNF-alpha ELISA
An enzyme-linked immunosorbent assay (ELISA) for Human Tumor Necrosis Factor-a
(TNF-a), Human Interferon-I3 (IFN-13) and Human Granulocyte-Colony Stimulating
Factor (G-CSF)
secreted from in vitro-transfected Human Keratinocyte cells is tested for the
detection of a cellular
innate immune response.
Cells are grown in EPILIFE medium with Human Growth Supplement in the absence
of
hydrocortisone from Invitrogen at a confluence of >70%. Cells are reverse
transfected with Ong,
93.75ng, 187.5ng, 375ng, 750ng, 150Ong or 3000ng of the indicated chemically
modified mRNA
- 221 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
complexed with RNAIMAXTm from Invitrogen as described in triplicate. Secreted
TNF-a in the culture
medium is measured 24 hours post-transfection for each of the chemically
modified mRNAs using an
ELISA kit from Invitrogen according to the manufacturer protocols.
Secreted IFN-I3 is measured 24 hours post-transfection for each of the
chemically modified
mRNAs using an ELISA kit from Invitrogen according to the manufacturer
protocols. Secreted hu-G-
CSF concentration is measured at 24 hours post-transfection for each of the
chemically modified
mRNAs. Secretion of the polypeptide of interest from transfected human cells
is quantified using an
ELISA kit from Invitrogen or R&D Systems (Minneapolis, MN) following the
manufacturers
recommended instructions. These data indicate which modified mRNA are capable
eliciting a reduced
cellular innate immune response in comparison to natural and other chemically
modified
polynucleotides or reference compounds by measuring exemplary type 1 cytokines
such as TNF-
alpha and IFN-beta.
Example 9. Cytotoxicity and Apoptosis
This experiment demonstrates cellular viability, cytotoxity and apoptosis for
distinct
modified mRNA-in vitro transfected Human Keratinocyte cells. Keratinocytes are
grown in EPILIFE
medium with Human Keratinocyte Growth Supplement in the absence of
hydrocortisone from
Invitrogen at a confluence of >70%. Keratinocytes are reverse transfected with
Ong, 46.875ng,
93.75ng, 187.5ng, 375ng, 750ng, 150Ong, 3000ng, or 6000ng of modified mRNA
complexed with
RNAIMAXTm from Invitrogen. The modified mRNA: RNAIMAXTm complex is formed.
Secreted huG-
CSF concentration in the culture medium is measured at 0, 6, 12, 24, and 48
hours post-transfection
for each concentration of each modified mRNA in triplicate. Secretion of the
polypeptide of interest
from transfected human keratinocytes is quantified using an ELISA kit from
Invitrogen or R&D
Systems following the manufacturers recommended instructions. Cellular
viability, cytotoxicity and
apoptosis is measured at 0, 12, 48, 96, and 192 hours post-transfection using
the APOTOX-GLOTm
kit from Promega (Madison, WI) according to manufacturer instructions.
Example 10. Incorporation of naturally and non-naturally occuring nucleosides
Naturally and non-naturally occurring nucleosides are incorporated into mRNA
encoding a
polypeptide of interest. Examples of these are given in Tables 4 and 5.
Certain commercially
available nucleoside triphosphates (NTPs) are investigated in the
polynucleotides of the invention. A
selection of these is given in Table 10. The resultant mRNAs are then examined
for their ability to
produce protein, induce cytokines, and/or produce a therapeutic outcome.
- 222 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 10: Naturally occurring nucleotide triphosphates.
Chemistry Modification Compound #
2-0-methylcytidine TP 00901074001
(1)
4-thiouridine TP 00901013011
(2)
2-0-methyluridine TP 00901073001
(3)
5-methyl-2-thiouridine TP 00901013003
(4)
5,2-0-dimethyluridine TP 03601073014
(5)
5-aminomethy1-2-thiouridine TP 00901013015
(6)
5,2-0-dimethylcytidine TP 00901074002
(7)
2-methylthio-N6-isopentenyladenosine TP 00901011015
(8)
2-0-methyladenosine TP 00901071001
(9)
2-0-methylguanosine TP 00901072001
(10)
N6-methyl-N6-threonylcarbamoyladenosine TP 03601011016
(11)
N6-hydroxynorvalylcarbamoyladenosine TP 00901011017
(12)
2-methylthio-N6-hydroxynorvalylcarbamoyladenosine TP 00901011018
(13)
2-0-ribosyladenosine (phosphate) TP 00901461001
(14)
N6,2-0-dimethyladenosine TP 00901071006
(15)
N6,N6,2-0-trimethyladenosine TP 00901071012
(16)
1,2-0-dimethyladenosine TP 00901071008
(17)
N6-acetyladenosine TP 00901011013
(18)
2-methyladenosine TP 00901011014
(19)
2-methylthio-N6-methyladenosine TP 00901011019
(20)
N2,2-0-dimethylguanosine TP 03601072014
(21)
N2,N2,2-0-trimethylguanosine TP 03601072015
(22)
7-cyano-7-deazaguanosine TP 03601012016
(23)
7-aminomethy1-7-deazaguanosine TP 03601012017
(24)
2-0-ribosylguanosine (phosphate) TP 00901462001
(25)
N2,7-dimethylguanosine TP 00901012018
(26)
- 223 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
N2,N2,7-trimethylguanosine TP 03601012019
(27)
1,2-0-dimethylguanosine TP 03601072008
(28)
Peroxywybutosine TP 00901012023
(29)
Hydroxywybutosine TP 00901012024
(30)
undermodified hydroxywybutosine TP 00901012025
(31)
Methylwyosine TP 00901012026
(32)
N2,7,2-0-trimethylguanosine TP 00901072018
(33)
1,2-0-dimethylinosine TP 00901072027
(34)
2-0-methylinosine TP 00901072028
(35)
4-demethylwyosine TP 00901012029
(36)
Isowyosine TP 00901012030
(37)
Queuosine TP 00901012031
(38)
Epoxyqueuosine TP 00901012032
(39)
galactosyl-queuosine TP 00901012033
(40)
mannosyl-queuosine TP 00901012034
(41)
Archaeosine TP 00901012035
(42)
Non-natural nucleotides of the present invention may also include those listed
below in
Table 11.
Table 11: Non-naturally occurring nucleotide triphosphates.
Chemistry Modification Compound #
036012293016
5-(1-Propynyl)ara-uridine TP
(43)
Z-0-Methyl-5-(1-propynyOuridine TP 03601073016 (44)
Z-0-Methyl-5-(1-propynyl)cytidine TP 03601074012 (45)
5-(1-Propynyl)ara-cytidine TP 03601294012 (46)
5-Ethynylara-cytidine TP 03601294011(47)
5-Ethynylcytidine TP 03601014011(48)
5-Vinylarauridine TP 03601013017 (49)
(Z)-5-(2-Bromo-vinyl)ara-uridine TP 03601293018 (50)
(E)-5-(2-Bromo-vinyl)ara-uridine TP 03601293019 (51)
(Z)-5-(2-Bromo-vinyl)uridine TP 03601013018 (52)
(E)-5-(2-Bromo-vinyl)uridine TP 03601013019 (53)
5-Methoxycytidine TP 03601014030 (54)
5-Formyluridine TP 03601013020 (55)
5-Cyanouridine TP 03601013021 (56)
- 224 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
5-Dimethylaminouridine TP 03601013022 (57)
5-Trideuteromethy1-6-deuterouridine TP 03601013023 (58)
5-Cyanocytidine TP 03601014031 (59)
5-(2-Chloro-phenyl)-2-thiocytidine TP 03601014032 (60)
5-(4-Amino-phenyl)-2-thiocytidine TP 03601014033 (61)
5-(2-Furanyl)uridine TP 03601013024 (62)
5-Phenylethynyluridine TP 03601013025 (63)
N4,2'-0-Dimethylcytidine TP 00901074004 (64)
3'-Ethynylcytidine TP 00901304001 (65)
4'-Carbocyclic adenosine TP 00901171001(66)
4'-Carbocyclic cytidine TP 00901174001(67)
4'-Carbocyclic guanosine TP 00901172001(68)
4'-Carbocyclic uridine TP 00901173001(69)
4'-Ethynyladenosine TP 00901311001(70)
4'-Ethynyluridine TP 00901313001(71)
4'-Ethynylcytidine TP 00901314001(72)
4'-Ethynylguanosine TP 00901312001 (73)
4'-Azidouridine TP 00901323001 (74)
4'-Azidocytidine TP 00901324001 (75)
4'-Azidoadenosine TP 0090132001(76)
4'-Azidoguanosine TP 00901322001 (77)
2'-Deoxy-2',2'-difluorocytidine TP 00901334001(78)
2'-Deoxy-2',2'-difluorouridine TP 00901333001(79)
2'-Deoxy-2',2'-difluoroadenosine TP 00901331001(80)
2'-Deoxy-2',2'-difluoroguanosine TP 00901332001(81)
2'-Deoxy-2'-b-fluorocytidine TP 00901024001(82)
2'-Deoxy-2'-b-fluorouridine TP 00901023001(83)
2'-Deoxy-2'-b-fluoroadenosine TP 00901021001(84)
2'-Deoxy-2'-b-fluoroguanosine TP 00901022001(85)
8-Trifluoromethyladenosine TP 03601011020 (86)
2'-Deoxy-2'-b-chlorouridine TP 00901033001(87)
2'-Deoxy-2'-b-bromouridine TP 00901043001(88)
2'-Deoxy-2'-b-iodouridine TP 00901053001(89)
2'-Deoxy-2'-b-chlorocytidine TP 00901034001(90)
2'-Deoxy-2'-b-bromocytidine TP 00901044001(91)
2'-Deoxy-2'-b-iodocytidine TP 00901054001(92)
2'-Deoxy-2'-b-chloroadenosine TP 00901031001(93)
2'-Deoxy-2'-b-bromoadenosine TP 00901041001(94)
2'-Deoxy-2'-b-iodoadenosine TP 00901051001(95)
2'-Deoxy-2'-b-chloroguanosine TP 00901032001(96)
2'-Deoxy-2'-b-bromoguanosine TP 00901042001(97)
2'-Deoxy-2'-b-iodoguanosine TP 00901052001(98)
5'-Homo-cytidine TP 00901344001 (99)
00901341001
5'-Homo-adenosine TP (100)
00901343001
5'-Homo-uridine TP (101)
00901342001
5'-Homo-guanosine TP (102)
- 225 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
00901353001
2'-Deoxy-2'-a-mercaptouridine TP (103)
00901363001
2'-Deoxy-2'-a-thiomethoxyuridine TP (104)
00901373001
2'-Deoxy-2'-a-azidouridine TP (105)
00901383001
2'-Deoxy-2'-a-aminouridine TP (106)
00901354001
2'-Deoxy-2'-a-mercaptocytidine TP (107)
00901364001
2'-Deoxy-2'-a-thiomethoxycytidine TP (108)
00901374001
2'-Deoxy-2'-a-azidocytidine TP (109)
00901384001
2'-Deoxy-2'-a-aminocytidine TP (110)
00901351001
2'-Deoxy-2'-a-mercaptoadenosine TP (111)
00901361001
2'-Deoxy-2'-a-thiomethoxyadenosine TP (112)
00901371001
2'-Deoxy-2'-a-azidoadenosine TP (113)
00901381001
2'-Deoxy-2'-a-aminoadenosine TP (114)
00901352001
2'-Deoxy-2'-a-mercaptoguanosine TP (115)
00901362001
2'-Deoxy-2'-a-thiomethoxyguanosine TP (116)
00901372001
2'-Deoxy-2'-a-azidoguanosine TP (117)
00901382001
2'-Deoxy-2'-a-aminoguanosine TP (118)
00901393001
2'-Deoxy-2'-b-mercaptouridine TP (119)
00901403001
2'-Deoxy-2'-b-thiomethoxyuridine TP (120)
00901413001
2'-Deoxy-2'-b-azidouridine TP (121)
00901423001
2'-Deoxy-2'-b-aminouridine TP (122)
00901394001
2'-Deoxy-2'-b-mercaptocytidine TP (123)
00901404001
2'-Deoxy-2'-b-thiomethoxycytidine TP (124)
00901414001
2'-Deoxy-2'-b-azidocytidine TP (125)
00901424001
2'-Deoxy-2'-b-aminocytidine TP (126)
00901391001
2'-Deoxy-2'-b-mercaptoadenosine TP (127)
00901401001
2'-Deoxy-2'-b-thiomethoxyadenosine TP (128)
00901411001
2'-Deoxy-2'-b-azidoadenosine TP (129)
2'-Deoxy-2'-b-aminoadenosine TP 00901421001(130)
00901392001
2'-Deoxy-2'-b-mercaptoguanosine TP (131)
- 226 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
00901402001
2'-Deoxy-2'-b-thiomethoxyguanosine TP (132)
00901412001
2'-Deoxy-2'-b-azidoguanosine TP (133)
00901422001
2'-Deoxy-2'-b-aminoguanosine TP (134)
00901431001
2'-b-Trifluoromethyladenosine TP (135)
00901434001
2'-b-Trifluoromethylcytidine TP (136)
00901432001
2'-b-Trifluoromethylguanosine TP (137)
00901433001
2'-b-Trifluoromethyluridine TP (138)
00901441001
2'-a-Trifluoromethyladenosine TP (139)
00901444001
2'-a-Trifluoromethylcytidine TP (140)
00901442001
2'-a-Trifluoromethylguanosine TP (141)
00901443001
2'-a-Trifluoromethyluridine TP (142)
2'-b-Ethynyladenosine TP 00901441001(143)
00901444001
2'-b-Ethynylcytidine TP (144)
2'-b-Ethynylguanosine TP 00901442001(145)
00901443001
2'-b-Ethynyluridine TP (146)
00901451001
2'-a-Ethynyladenosine TP (147)
00901454001
2'-a-Ethynylcytidine TP (148)
00901452001
2'-a-Ethynylguanosine TP (149)
00901453001
2'-a-Ethynyluridine TP (150)
03601014034
(E)-5-(2-Bromo-vinyl)cytidine TP (151)
03601011021
2-Trifluoromethyladenosine TP (152)
03601011022
2-Mercaptoadenosine TP (153)
03601011002
2-Aminoadenosine TP (154)
03601011023
2-Azidoadenosine TP (155)
03601011024
2-Fluoroadenosine TP (156)
03601011025
2-Chloroadenosine TP (157)
03601011026
2-Bromoadenosine TP (158)
03601011027
2-lodoadenosine TP (159)
03601011038
Formycin A TP (160)
Formycin B TP 03601011039
- 227 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(161)
03601011040
Oxoformycin TP (162)
03601011037
Pyrrolosine TP (163)
03601011028
9-Deazaadenosine TP (164)
03601012020
9-Deazaguanosine TP (165)
03601011029
3-Deazaadenosine TP (166)
03601011030
3-Deaza-3-fluoroadenosine TP (167)
03601011031
3-Deaza-3-chloroadenosine TP (168)
03601011032
3-Deaza-3-bromoadenosine TP (169)
03601011033
3-Deaza-3-iodoadenosine TP (170)
03601011034
1-Deazaadenosine TP (171)
Example 11. Directed SAR of Pseudouridine and N1-methyl PseudoUridine
With the recent focus on the pyrimidine nucleoside pseudouridine, a series of
structure-
activity studies were designed to investigate mRNA containing modifications to
pseudouridine or N1-
methyl-pseudourdine.
The study was designed to explore the effect of chain length, increased
lipophilicity,
presence of ring structures, and alteration of hydrophobic or hydrophilic
interactions when
modifications were made at the Ni position, C6 position, the 2-position, the 4-
position and on the
phosphate backbone. Stability is also investigated.
To this end, modifications involving alkylation, cycloalkylation, alkyl-
cycloalkylation,
arylation, alkyl-arylation, alkylation moieties with amino groups, alkylation
moieties with carboxylic
acid groups, and alkylation moieties containing amino acid charged moieties
are investigated. The
degree of alkylation is generally C1-C6. Examples of the chemistry
modifications include those listed
in Tables 12, 13 and 14.
- 228 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 12: Pseudouridine and N1-methyl Pseudo Uridine SAR.
Chemistry Modification Compound # Naturally
occuring
N1-Modifications
1-Ethyl-pseudo-UTP 03601015003 N
(172)
1-Propyl-pseudo-UTP 03601015004 N
(173)
1-iso-propyl-pseudo-UTP 03601015028(174) N
1-(2,2,2-Trifluoroethyl)-pseudo-UTP 03601015005 N
(175)
1-Cyclopropyl-pseudo-UTP 03601015029(176) N
1-Cyclopropylmethyl-pseudo-UTP 03601015030(177) N
1-Phenyl-pseudo-UTP 03601015031 N
(178)
1-Benzyl-pseudo-UTP 03601015032 N
(179)
1-Aminomethyl-pseudo-UTP 03601015033 N
(180)
Pseudo-UTP-1-2-ethanoic acid 03601015034 N
(181)
1-(3-Amino-3-carboxypropyl)pseudo-UTP 03601015035 N
(182)
1-Methyl-3-(3-amino-3-carboxypropyl)pseudo-UTP 03601015036 Y
(183)
C-6 Modifications
6-Methyl-pseudo-UTP 03601015037 N
(184)
6-Trifluoromethyl-pseudo-UTP 03601015038 N
(185)
6-Methoxy-pseudo-UTP 03601015039 N
(186)
6-Phenyl-pseudo-UTP 03601015040 N
(187)
6-lodo-pseudo-UTP 03601015041 N
(188)
6-Bromo-pseudo-UTP 03601015042 N
(189)
6-Chloro-pseudo-UTP 03601015043 N
(190)
6-Fluoro-pseudo-UTP 03601015044 N
(191)
2- or 4-position Modifications
4-Thio-pseudo-UTP 00901015022 N
(192)
2-Thio-pseudo-UTP 00901015006 N
(193)
Phosphate backbone Modifications
Alpha-thio-pseudo-UTP 00902015001 N
(194)
1-Me-alpha-thio-pseudo-UTP 00902015002 N
(195)
- 229 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 13: Pseudouridine and N1-methyl Pseudo Uridine SAR.
Chemistry Modification Compound # Naturally
occuring
1-Methyl-pseudo-UTP 00901015002 Y
(196)
1-Butyl-pseudo-UTP 03601015045 N
(197)
1-tert-Butyl-pseudo-UTP 03601015046 N
(198)
1-Pentyl-pseudo-UTP 03601015047 N
(199)
1-Hexyl-pseudo-UTP 03601015048 N
(200)
1-Trifluoromethyl-pseudo-UTP 03601015049 Y
(201)
1-Cyclobutyl-pseudo-UTP 03601015050 N
(202)
1-Cyclopentyl-pseudo-UTP 03601015051 N
(203)
1-Cyclohexyl-pseudo-UTP 03601015052 N
(204)
1-Cycloheptyl-pseudo-UTP 03601015053 N
(205)
1-Cyclooctyl-pseudo-UTP 03601015054 N
(206)
1-Cyclobutylmethyl-pseudo-UTP 03601015055 N
(207)
1-Cyclopentylmethyl-pseudo-UTP 03601015056 N
(208)
1-Cyclohexylmethyl-pseudo-UTP 03601015057 N
(209)
1-Cycloheptylmethyl-pseudo-UTP 03601015058 N
(210)
1-Cyclooctylmethyl-pseudo-UTP 03601015059 N
(211)
1-p-tolyl-pseudo-UTP 03601015060 N
(212)
1-(2,4,6-Trinnethyl-phenyl)pseudo-UTP 03601015061 N
(213)
1-(4-Methoxy-phenyl)pseudo-UTP 03601015062 N
(214)
1-(4-Amino-phenyl)pseudo-UTP 03601015063 N
(215)
1(4-Nitro-phenyl)pseudo-UTP 03601015064 N
(216)
Pseudo-UTP-N1-p-benzoic acid 03601015065 N
(217)
1-(4-Methyl-benzyl)pseudo-UTP 03601015066 N
(218)
1-(2,4,6-Trimethyl-benzyl)pseudo-UTP 03601015067 N
(219)
1-(4-Methoxy-benzyl)pseudo-UTP 03601015068 N
(220)
1-(4-Amino-benzyl)pseudo-UTP 03601015069 N
(221)
1-(4-Nitro-benzyl)pseudo-UTP 03601015070 N
(222)
- 230 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Pseudo-UTP-N1-methyl-p-benzoic acid 03601015071 N
(223)
1-(2-Amino-ethyl)pseudo-UTP 03601015072 N
(224)
1-(3-Amino-propyl)pseudo-UTP 03601015073 N
(225)
1-(4-Amino-butyl)pseudo-UTP 03601015074 N
(226)
1-(5-Amino-pentyl)pseudo-UTP 03601015075 N
(227)
1-(6-Amino-hexyl)pseudo-UTP 03601015076 N
(228)
Pseudo-UTP-N1-3-propionic acid 03601015077 N
(229)
Pseudo-UTP-N1-4-butanoic acid 03601015078 N
(230)
Pseudo-UTP-N1-5-pentanoic acid 03601015079 N
(231)
Pseudo-UTP-N1-6-hexanoic acid 03601015080 N
(232)
Pseudo-UTP-N1-7-heptanoic acid 03601015081 N
(233)
1-(2-Amino-2-carboxyethyl)pseudo-UTP 03601015082 N
(234)
1-(4-Amino-4-carboxybutyl)pseudo-UTP 03601015083 N
(235)
3-Alkyl-pseudo-UTP 00901015187 N
(236)
6-Ethyl-pseudo-UTP 03601015084 N
(237)
6-Propyl-pseudo-UTP 03601015085 N
(2380
6-iso-Propyl-pseudo-UTP 03601015086 N
(239)
6-Butyl-pseudo-UTP 03601015087 N
(240)
6-tert-Butyl-pseudo-UTP 03601015088 N
(241)
6-(2,2,2-Trifluoroethyl)-pseudo-UTP 03601015089 N
(242)
6-Ethoxy-pseudo-UTP 03601015090 N
(243)
6-Trifluoromethoxy-pseudo-UTP 03601015091 N
(244)
6-Phenyl-pseudo-UTP 03601015092 N
(245)
6-(Substituted-Phenyl)-pseudo-UTP 03601015093 N
(246)
6-Cyano-pseudo-UTP 03601015094 N
(247)
6-Azido-pseudo-UTP 03601015095 N
(248)
6-Amino-pseudo-UTP 03601015096 N
(249)
6-Ethylcarboxylate-pseudo-UTP 03601015097 N
(250)
6-Hydroxy-pseudo-UTP 03601015098 N
(251)
- 231 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
6-Methylamino-pseudo-UTP 03601015099 N
(252)
6-Dimethylamino-pseudo-UTP 03601015100 N
(253)
6-Hydroxyamino-pseudo-UTP 03601015101 N
(254)
6-Formyl-pseudo-UTP 03601015102 N
(255)
6-(4-Morpholino)-pseudo-UTP 03601015103 N
(256)
6-(4-Thiomorpholino)-pseudo-UTP 03601015104 N
(257)
1-Me-4-thio-pseudo-UTP 03601015105 N
(258)
1-Me-2-thio-pseudo-UTP 03601015106 N
(259)
1,6-Dimethyl-pseudo-UTP 03601015107 N
(260)
1-Methy1-6-trifluoromethyl-pseudo-UTP 03601015108 N
(261)
1-Methy1-6-ethyl-pseudo-UTP 03601015109 N
(262)
1-Methyl-6-propyl-pseudo-UTP 03601015110 N
(263)
1-Methy1-6-iso-propyl-pseudo-UTP 03601015111 0 N
(264)
1-Methyl-6-butyl-pseudo-UTP 03601015112 N
(265)
1-Methy1-6-tert-butyl-pseudo-UTP 03601015113 N
(266)
1-Methy1-6-(2,2,2-TrifluoroethyD 03601015114pseudo-UTP N
(267)
1-Methyl-6-iodo-pseudo-UTP 03601015115 N
(268)
1-Methyl-6-bromo-pseudo-UTP 03601015116 N
(269)
1-Methyl-6-chloro-pseudo-UTP 03601015117 N
(270)
1-Methyl-6-fluoro-pseudo-UTP 03601015118 N
(271)
1-Methyl-6-methoxy-pseudo-UTP 03601015119 N
(272)
1-Methy1-6-ethoxy-pseudo-UTP 03601015120 N
(273)
1-Methy1-6-trifluoromethoxy-pseudo-UTP 03601015121 N
(274)
1-Methy1-6-phenyl-pseudo-UTP 03601015122 N
(275)
1-Methy1-6-(substituted phenyl)pseudo-UTP 03601015123 N
(276)
1-Methy1-6-cyano-pseudo-UTP 03601015124 N
(277)
1-Methy1-6-azido-pseudo-UTP 03601015125 N
(278)
1-Methy1-6-amino-pseudo-UTP 03601015126 N
(279)
1-Methy1-6-ethylcarboxylate-pseudo-UTP 03601015127 N
(280)
- 232 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
03601015128
1-Methy1-6-hydroxy-pseudo-UTP
(281)
03601015129
1-Methy1-6-methylamino-pseudo-UTP
(282)
03601015130
1-Methy1-6-dimethylamino-pseudo-UTP
(283)
03601015131
1-Methy1-6-hydroxyamino-pseudo-UTP
(284)
03601015132
1-Methy1-6-formyl-pseudo-UTP
(285)
03601015133
1-Methy1-6-(4-morpholino)-pseudo-UTP
(286)
03601015134
1-Methy1-6-(4-thiomorpholino)-pseudo-UTP
(287)
03601015188
1-Alky1-6-vinyl-pseudo-UTP
(288)
03601015189
1-Alky1-6-allyl-pseudo-UTP
(289)
03601015190
1-Alky1-6-homoallyl-pseudo-UTP
(290)
03601015191
1-Alky1-6-ethynyl-pseudo-UTP
(291)
03601015192
1-Alky1-6-(2-propyny1)-pseudo-UTP
(292)
03601015193
1-Alky1-6-(1-propyny1)-pseudo-UTP
(293)
Additional non-naturally occurring compounds were designed for structure
activity
relationship around 1-methylpseudouridine. These compounds include those
listed in Table 14.
Table 14: Non-naturally occurring nucleotides designed using SAR around 1-
methylpseudouridine.
Chemistry Modification Compound #
03601015135
1-Hydroxymethylpseudouridine TP (294)
03601015136
1-(2-Hydroxyethyl)pseudouridine TP (295)
03601015137
1-Methoxymethylpseudouridine TP (296)
03601015138
1-(2-Methoxyethyl)pseudouridine TP (297)
03601015139
1-(2,2-DiethoxyethyDpseudouridine TP (298)
03601015140
( )1-(2-Hydroxypropyl)pseudouridine TP (299)
03601015141
(2R)-1-(2-Hydroxypropyl)pseudouridine TP (300)
03601015142
(2S)-1-(2-Hydroxypropyl)pseudouridine TP (301)
03601015143
1-Cyanomethylpseudouridine TP (302)
03601015144
1-Morpholinomethylpseudouridine TP (303)
03601015145
1-Thiomorpholinomethylpseudouridine TP (304)
03601015146
1-Benzyloxymethylpseudouridine TP (305)
1-(2,2,3,3,3-Pentafluoropropyl)pseudouridine TP 03601015147
- 233 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
(306)
03601015148
1-Thiomethoxymethylpseudouridine TP (307)
03601015149
1-Methanesulfonylmethylpseudouridine TP (308)
03601015150
1-Vinylpseudouridine TP (309)
03601015151
1-Allylpseudouridine TP (310)
03601015152
1-Homoallylpseudouridine TP (311)
03601015153
1-Propargylpseudouridine TP (312)
03601015154
1-(4-Fluorobenzyl)pseudouridine TP (313)
03601015155
1-(4-Chlorobenzyl)pseudouridine TP (314)
03601015156
1-(4-Bromobenzyl)pseudouridine TP (315)
03601015157
1-(4-lodobenzyl)pseudouridine TP (316)
03601015158
1-(4-Methylbenzyl)pseudouridine TP (317)
03601015159
1-(4-Trifluoromethylbenzyl)pseudouridine TP (318)
03601015160
1-(4-Methoxybenzyl)pseudouridine TP (319)
03601015161
1-(4-Trifluoromethoxybenzyl)pseudouridine TP (320)
03601015162
1-(4-Thiomethoxybenzyl)pseudouridine TP (321)
03601015163
1-(4-Methanesulfonylbenzyl)pseudouridine TP (322)
03601015164
Pseudouridine 1-(4-methylbenzoic acid) TP (323)
03601015165
Pseudouridine 1-(4-methylbenzenesulfonic acid) TP (324)
03601015166
1-(2,4,6-Trimethylbenzyl)pseudouridine TP (325)
03601015167
1-(4-Nitrobenzyl)pseudouridine TP (326)
03601015168
1-(4-Azidobenzyl)pseudouridine TP (327)
03601015169
1-(3,4-Dimethoxybenzyl)pseudouridine TP (328)
03601015170
1-(3,4-Bis-trifluoromethoxybenzyl)pseudouridine TP (329)
03601015171
1-Acetylpseudouridine TP (330)
03601015172
1-Trifluoroacetylpseudouridine TP (331)
03601015173
1-Benzoylpseudouridine TP (332)
03601015174
1-Pivaloylpseudouridine TP (333)
03601015175
1-(3-Cyclopropyl-prop-2-ynyl)pseudouridine TP (334)
Pseudouridine TP 1-methylphosphonic acid diethyl ester 03601015176
- 234 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(335)
03601015177
Pseudouridine TP 1-methylphosphonic acid (336)
03601015178
Pseudouridine TP 1-[3-(2-ethoxy)]propionic acid (337)
Pseudouridine TP 143-{2-(2-ethoxy)-ethoxy}] propionic 03601015179
acid (338)
Pseudouridine TP 143-{2-(242-ethoxy ]-ethoxy)- 03601015180
ethoxy}]propionic acid (339)
Pseudouridine TP 143-{2-(242-(2-ethoxy )-ethoxy]-ethoxy 03601015181
)-ethoxy}]propionic acid (340)
Pseudouridine TP 1-[3-{2-(2-[2-{2(2-ethoxy )-ethoxy}- 03601015182
ethoxy]-ethoxy )-ethoxy}]propionic acid (341)
1-{342-(2-Aminoethoxy)-ethoxyypropionyl } 03601015183
pseudouridine TP (342)
143-(2-{242-(2-Aminoethoxy)-ethoxyyethoxy}-ethoxy)- 03601015184
propionyl]pseudouridine TP (343)
03601015185
1-Biotinylpseudouridine TP (344)
03601015186
1-Biotinyl-PEG2-pseudouridine TP (345)
Example 12. Incorporation of naturally and non-naturally occuring nucleosides
Naturally and non-naturally occurring nucleosides are incorporated into mRNA
encoding a
polypeptide of interest. Examples of these are given in Tables 15 and 16.
Certain commercially
available nucleoside triphosphates (NTPs) are investigated in the
polynucleotides of the invention. A
selection of these are given in Table 15. The resultant mRNA are then examined
for their ability to
produce protein, induce cytokines, and/or produce a therapeutic outcome.
Table 15: Naturally and non-naturally occurring nucleotides.
Naturally
Chemistry Modification Compound #
occuring
N4-Methyl-Cytidine TP 00901014004
(346)
N4,N4-Dimethy1-2'-0Me-Cytidine TP 03601014029
(347)
00901013004
5-Oxyacetic acid-methyl ester-Uridine TP
(348)
00901015007
3-Methyl-pseudo-Uridine TP
(349)
00901014005
5-Hydroxymethyl-Cytidine TP
(350)
00901014003
5-Trifluoromethyl-Cytidine TP
(3510
00901013002
5-Trifluoromethyl-Uridine TP
(352)
00901013006
5-Methyl-amino-methyl-Uridine TP
(353)
00901013026
5-Carboxy-methyl-amino-methyl-Uridine TP
(354)
5-Carboxymethylaminomethy1-2'-0Me-Uridine TP 00901023026
(355)
5-Carboxymethylaminomethy1-2-thio-Uridine TP 00901013027
- 235 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(356)
5-Methylaminomethy1-2-thio-Uridine TP 00901013028
(357)
5-Methoxy-carbonyl-methyl-Uridine TP 00901013005
(358)
5-Methoxy-carbonyl-methyl-2'-0Me-Uridine TP 00901023005
(359)
5-Oxyacetic acid- Uridine TP 00901013029
(360)
3-(3-Amino-3-carboxypropyI)-Uridine TP 00901013030
(361)
5-(carboxyhydroxymethyl)uridine methyl ester TP 00901013031
(3620
5-(carboxyhydroxymethyl)uridine TP 00901013032
(363)
Table 16: Non-naturally occurring nucleoside triphosphates.
Chemistry Modification Compound #
1-Me-GTP 00901012008
(364)
2'-0Me-2-Amino-ATP 00901071002
(365)
2'-0Me-pseudo-UTP 00901075001
(366)
2'-0Me-6-Me-UTP 03601073033
(367)
2'-Azido-2'-deoxy-ATP 00901371001
(368)
2'-Azido-2'-deoxy-GTP 00901372001
(369)
2'-Azido-2'-deoxy-UTP 00901373001
(370)
2'-Azido-2'-deoxy-CTP 00901374001
(371)
2'-Amino-2'-deoxy-ATP 00901381001
(372)
2'-Amino-2'-deoxy-GTP 00901382001
(373)
2'-Amino-2'-deoxy-UTP 00901383001
(374)
2'-Amino-2'-deoxy-CTP 00901384001
(375)
0
2-Amino-ATP 0901011002
(376)
8-Aza-ATP 00901011003
(377)
Xanthosine-5'-TP 00901012003
(378)
0
5-Bromo-CTP 3601014008
(379)
2'-F-5-Methyl-2'-deoxy-UTP 03601023014
(380)
5-Aminoallyl-CTP 03601014009
(381)
2-Amino-riboside-TP 03601012004
(382)
- 236 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 13. Incorporation of modifications to the nucleobase and carbohydrate
(sugar)
Naturally and non-naturally occurring nucleosides are incorporated into mRNA
encoding a
polypeptide of interest. Commercially available nucleosides and NTPs having
modifications to both
the nucleobase and carbohydrate (sugar) are examined for their ability to be
incorporated into mRNA
and to produce protein, induce cytokines, and/or produce a therapeutic
outcome. Examples of these
nucleosides are given in Tables 17 and 18.
Table 17: Selected modifications.
Chemistry Modification Compound #
03601023034
5-iodo-2'-fluoro-deoxyuridine TP
(383)
00901014035
5-iodo-cytidine TP
(384)
00901043001
2'-bromo-deoxyuridine TP
(385)
03601011035
8-bromo-adenosine TP
(386)
03601012021
8-bromo-guanosine TP
(387)
00901144001
2,2'-anhydro-cytidine TP hydrochloride
(388)
00901143001
2,2'-anhydro-uridine TP
(389)
00901373001
2'-Azido-deoxyuridine TP
(390)
03601011002
2-amino-adenosine TP
(391)
03601014013
N4-Benzoyl-cytidine TP
(392)
03601014037
N4-Amino-cytidine TP
(393)
00901074007
2'-0-Methyl-N4-Acetyl-cytidine TP
(394)
00901024007
2'Fluoro-N4-Acetyl-cytidine TP
(395)
03601024013
2'Fluor-N4-Bz-cytidine TP
(396)
03601074013
2'0-methyl-N4-Bz-cytidine TP
(397)
03601071036
2'0-methyl-N6-Bz-deoxyadenosine TP
(398)
03601021036
2'Fluoro-N6-Bz-deoxyadenosine TP
(399)
03601012022
N2-isobutyl-guanosine TP
(400)
03601022022
2'Fluro-N2-isobutyl-guanosine TP
(401)
03601072022
2'0-methyl-N2-isobutyl-guanosine TP
(402)
- 237 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Table 18: Selected combinations.
Name Compound #
00901013035
5-Methoxycarbonylmethy1-2-thiouridine TP
(403)
00901013028
5-Methylaminomethy1-2-thiouridine TP
(404)
00901013036
5-Carbamoylmethyluridine TP
(405)
00901073036
5-Carbamoylmethyl-2-0-methyluridine TP
(406)
1-Methy1-3-(3-amino-3-carboxypropyl) pseudouridine 00901015036
TP (407)
00901013037
5-Methylaminomethy1-2-selenouridine TP
(408)
00901013038
5-Carboxymethyluridine TP
(409)
03601013039
5-Methyldihydrouridine TP
(410)
00901014038
lysidine TP
(411)
00901013040
5-Taurinomethyluridine TP
(412)
00901013041
5-Taurinomethy1-2-thiouridine TP
(413)
00901013042
5-(iso-PentenylaminomethyOuridine TP
(414)
00901013043
5-(iso-Pentenylaminomethyl)- 2-thiouridine TP
(415)
00901013044
5-(iso-Pentenylaminomethyl)-2-0-methyluridine TP
(416)
00901074007
N4-Acetyl-2-0-methylcytidine TP
(417)
00901074004
N4,2-O-Dimethylcytidine TP
(418)
03601074036
5-Formyl-2-0-methylcytidine TP
(419)
00901073001
2-0-Methylpseudouridine TP
(420)
00901073008
2-Thio-2-0-methyluridine TP
(421)
00901073045
3,2-0-Dimethyluridine TP
(422)
In the tables "UTP" stands for uridine triphosphate, "GTP" stands for
guanosine
triphosphate, "ATP" stands for adenosine triphosphate, "CTP" stands for
cytosine triphosphate, "TP"
stands for triphosphate and "Bz" stands for benzoyl.
The non-naturally occurring nucleobases of the invention, e.g., as indicated
in Tables 5-
10, can be provided as the 5'-mono-, di-, or triphosphate and/or the 3'-
phosphoramidite (e.g., the 2-
cyanoethyl-N,N-diisopropylphosphoramidite).
- 238 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 14. Synthesis of pseudo-U-alpha-thio-TP (00902015001 (194))
Scheme 2
HN ANH HN ANH
HO- 0 CI-P-0-
1. Trimethyl phosphate
4. Bu3N
2. Proton Sponge
OH OH OH OH
5. Bis(tributylammonium)
1 3. PSCI3
2
pyrophosphate
C31-112N206 6. ACN
Exact Mass: 244.07
HN ANH HN ANH
o, pH
S µ,
H H 7. 0.2 M TEAB P-0 S
HO -P-O-P-O-P-0- o 0 1:'/- 0
OH OH OH - P-0
0' \
011
OH OH OH OH
4 3
C31-115N2014P35
Exact Mass: 499.95 C31-113N2013P3S
Exact Mass: 481.94
A solution of pseudouridine 1 (130.0 mg, 0.53 mmol; applied heat to make it
soluble) and proton
sponge (170.4 mg, 0.8 mmol, 1.5 equiv.) in trimethyl phosphate (0.8 mL) was
stirred for 10.0 minutes
at 0 C. Thiophosphoryl chloride (107.5 pL, 1.06 mmol, 2.0 equiv.) was added
dropwise to the
solution and it was then kept stirring for 2.0 hours under N2 atmosphere. A
mixture of tributylamine
(514.84 pL, 2.13 mmol, 4.0 equiv.) and bis(tributylammonium) pyrophosphate
(872.4 mg, 1.59 mmol,
3.0 equiv.) in acetonitrile (2.5 mL) was added at once. After -25 minutes, the
reaction was quenched
with 24.5 mL of water and the clear solution was stirred vigorously for about
an hour at room
temperature. The pH of the solution was adjusted to 6.75 by adding 4.5 mL of
1.0 M TEAB buffer
along with vigorous stirring for about 3.0 hours. LCMS analysis indicated the
formation of the
corresponding triphosphate. The reaction mixture was then lyophilized
overnight. The crude reaction
mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative column, 250 x
30.0 mm, 5.0
micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A = 100 mM TEAB
buffer, B = ACN; flow
rate: 20.0 mL/min; retention time: 16.57-18.15 min). Fractions containing the
desired were pooled and
lyophilized to yield the Pseudo-U-alpha-thio-TP as a tetrakis(triethylammonium
salt) (62.73 mg, 24.5
%, based on a265 = 7,546). UVmax = 265 nm; MS: m/e 498.70 (M-H).
- 239 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 15. Synthesis of 1-methyl-pseudo-U-alpha-thio-TP (00902015002 (195))
Scheme 3
A A
HN N,Me HN N,Me
HO- CI-P-0-
1. Trimethyl phosphate
2. Proton Sponge CI
4. Bu3N
OH OH OH OH 5.
Bis(tributylammonium)
3. PSCI3
6
pyrophosphate
C10H14N206 6. ACN
Exact Mass: 258.09
A ,Me
HN N,Me HNA N
0, OH
0 0 S P/-0 S
H 7Ø2 M TEAB
HO-P-O-P-O-P-0- o 0 P/- 0 -
OH OH OH P. 0-
\
OH
OH OH OH OH
8 7
C10H17N2014P35 C10H15N2013P35
Exact Mass: 513.96 Exact Mass: 495.95
A solution of 1-methyl-pseudouridine 5 (130.0 mg, 0.5 mmol; applied heat to
make it soluble) and
5 proton sponge (160.7 mg, 0.75 mmol, 1.5 equiv.) in trimethyl phosphate
(0.8 mL) was stirred for 10.0
minutes at 0 C. Thiophosphoryl chloride (101.43 pL, 1.00 mmol, 2.0 equiv.)
was added dropwise to
the solution and it was then kept stirring for 2.0 hours under N2 atmosphere.
A mixture of
tributylamine (485.7 pL, 2.00 mmol, 4.0 equiv.) and bis(tributylammonium)
pyrophosphate (823.0 mg,
1.5 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once. After -25
minutes, the reaction was
quenched with 24.0 mL of water and the clear solution was stirred vigorously
for about an hour at
room temperature. The pH of the solution was adjusted to 6.85 by adding about
3.5 mL of 1.0 M
TEAB buffer along with vigorous stirring for about 3.0 hours. LCMS analysis
indicated the formation of
the corresponding triphosphate. The reaction mixture was then lyophilized
overnight. The crude
reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x 30.0
mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A = 100 mM
TEAB buffer, B =
ACN; flow rate: 20.0 mL/min; retention time: 17.34-18.72 min). Fractions
containing the desired were
pooled and lyophilized to yield the 1-Methyl-Pseudo-U-alpha-thio-TP as a
tetrakis(triethylammonium
salt) (72.37 mg, 28.0 %, based on a271 = 8,500). UVmax = 271 nm; MS: m/e
512.66 (M-H).
- 240 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 16. Synthesis of 1-ethyl-pseudo-UTP (03601015003 (172))
Scheme 4
0
0 0
A
HNNH
FIN A NH HN AN
HO
Ac20, DM F
DMAP Ac o 1 BSA
0
Ac0
-30 C, V0 2 I-CH2CH3
HO
AcO -0Ac
1 Ac0 oAc
9
0
HN
NH3/Me0H HO
____________ >
HO
Compound 9: To a solution of pseudouridine (1, 2.4 g, 9.8 mmol) in anhydrous
N,N-
dimethylformamide (30 mL) at -30 C was added 4-dimethylaminopyridine (DMAP,
1.1 g, 9.8 mmol),
5 followed by acetic anhydride (10 mL) portion wise over a period of 15
min. The reaction mixture was
stirred at -30 C for 3 h, and then the temperature was raised to room
temperature. The reaction
mixture was quenched with Me0H (10 mL), and concentrated to dryness under
reduced pressure.
The residue was dissolved in CH2C12 (100 mL), and washed with H20 (50 mL). The
organic phase
was dried (Na2SO4) and concentrated. Then the crude compound 9 was dried
overnight in a vacuum
10 oven with P205 and used without further purification.
Compound 10: To a solution of 2',3',5'-tri-O-acetyl-pseudo uridine (9) (0.8
g,2.2 mmol) in dry CH3CN
(20 mL) was added N,0-bis(trimethylsily1)acetamide (BSA) (3.0 mL), and the
reaction mixture was
reflux for 2 h. The reaction mixture was then cooled to room temperature.
CH3CH2I (0.5 g, 3.3 mmol)
was added, and the reaction mixture was stirred at 62 C overnight. Then
CH3CH2I (0.5 g, 3.3 mmol)
was added, and the reaction mixture was stirred at 62 C for four days. The
reaction mixture was
evaporated under reduced pressure. The residue was dissolved in CH2C12 (100
mL), washed with 1%
NaHCO3 solution (50 mL), dried (Na2SO4) and evaporated to dryness. The
residual was purified by
silica gel column using PE: EA (5:1 to 1:1) as the eluent to give 0.56 g of
desired product 10.
1-Ethyl-pseudouridine 11: A solution of compound 10 (0.56 g) in ammonia
saturated methanol (50
mL) was stirred at room temperature overnight. The volatiles were removed
under reduced pressure.
Then the residue was purified by silica gel column chromatography, eluted with
5-10% methanol in
dichloromethane to give 230 mg compound 11 as a light yellow solid with 95.95%
HPLC purity. 1H-
NMR (DMSO-d6, 300 MHz, ppm) 6 11.32 (br, 1H), 7.81 (s, 1H), 5.01 (d, J=
3.00Hz, 1H), 4.98 (t, J=
3.00Hz, 1H), 4.75 (dd, J= 1.5, 2.7 Hz, 1H), 4.46 (d , J= 3.00Hz, 1H), 3.88-
3.95 (m, 2H), 3.69-3.70 (m,
4H), 3.45-3.48 (m, 1H), 1.17 (t, J =5.10Hz, 1H).
- 241 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Scheme 5
0 0
HNNC..3
/ HN ANCH3
O 9
HO -Q CI-P-0
1. Trimethyl phosphate CI
4. Bu3N
2. Proton Sponge
OH OH OH OH
5. Bis(tributylammonium)
11 3. POCI3
12 pyrophosphate
C11H16N206 6. ACN
Exact Mass: 272.10
0 0
HNNCH3 HNNCH3
0, pH
9 9 9 7. 0.2 M TEAB p
/13-0
OH OH OH -P-0
\
OH
OH OH OH OH
14
C11H19N2015P3 13
Exact Mass: 512.00
C11H17N2014P3
Exact Mass: 493.99
1-Ethyl-pseudo-UTP: A solution of 1-ethyl-pseudouridine 11 (124.0 mg, 0.46
mmol; applied heat to
make it soluble) and proton sponge (147.87 mg, 0.69 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8
mL) was stirred for 10.0 minutes at 0 c. Phosphorus oxychloride (85.9 pL,
0.92 mmol, 2.0 equiv.)
was added dropwise to the solution and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (446.5 pL, 1.8 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(757.2 mg, 1.38 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 25.0 mL of water and the clear solution was stirred
vigorously for about
an hour at room temperature. The pH of the solution was adjusted to 6.50 by
adding about 3.5 mL of
1.0 M TEAB buffer along with vigorous stirring for about 3.0 hours. LCMS
analysis indicated the
formation of the corresponding triphosphate. The reaction mixture was then
lyophilized overnight. The
crude reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x
30.0 mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A =
100 mM TEAB buffer,
B = ACN; flow rate: 20.0 mL/min; retention time: 17.87-18.68 min). Fractions
containing the desired
were pooled and lyophilized to yield the 1-Ethyl-pseudo-UTP as a
tetrakis(triethylammonium salt)
(47.7 mg, 20.2%, based on a271 = 8,500). UVmax = 271 nm; MS: m/e 510.70 (M-H).
- 242 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 17. Synthesis of 1-propyl-pseudo-UTP (03601015004 (173))
Scheme 6
0
0 0
HN A NH
HN A NH
AN
HO 0 -30 Ac20,DMAP DMF Ac 0 I-CH2CH2CH3
C,
DBU 0
Ha OH ________________________________________ = Ac0
Acas -0Ac
1 Aca 0Ac
9
0
HN AN
NH3/Me0H HO
Ha OH
16
Compound 15: To a solution of 2',3',5'-tri- 0-acetyl pseudouridine 9 (1.0 g,
2.7 mmol) in dry pyridine
(20 mL) was added DBU (0.6 g, 4.1 mmol), and the reaction mixture was stirred
at room temperature
5 for 0.5 h. To this mixture, CH3CH2CH2I (0.69 g, 4.0 mmol) was added and
stirred at room temperature
for 2-3 h. The reaction mixture was dissolved in CH2Cl2 (100 mL), washed with
brine (3 x 50 mL),
dried (Na2SO4) and evaporated to dryness. The residual was purified with
silica gel column using
PE:EA-10:1 to 3:1 as the eluent to afford 0.5 g desired compound 15.
10 1-
Propyl-pseudo-U (16): A solution of compound 15(0.5 g) in ammonia saturated
methanol (50 mL)
was stirred at room temperature overnight. The volatiles were removed under
reduced pressure. The
residue was purified by silica gel column chromatography, eluted with 5-10%
methanol in
dichloromethane to give 260 mg compound 16 as off-white solid with 96.59% HPLC
purity. Analytical
data for 1-Propyl-pseudo-U (16): 1H-NMR (DMSO-d6, 300 MHz, ppm) 6 11.29 (br,
1H), 7.79 (s, 1H),
15 4.96
(d, J =1.80Hz, 1H), 4.83 (t, J = 3.90Hz, 1H), 4.73 (d, J = 3.90Hz, 1H), 4.44
(d, J = 3.00Hz, 1H),
3.85-3.92 (m, 2H), 3.43-3.69 (m, 5H), 1.56 (q, J = 5.40Hz, 2H), 8.38 (t, J =
5.40Hz, 3H).
- 243 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Scheme 7
0 0
HN N
z/CH3 HN A N//CH3
C) 0
HO- CI-P-0-
0
1. Trimethyl phosphate
2. Proton Sponge CI
4. Bu3N
OH OH OH OH 5. Bis(tributylammonium)
16 3. POCI3 17
pyrophosphate
C12H18N206 6. ACN
Exact Mass: 286.12
0 0
HN N
z/CH3 HN NA //CH3
0, OH
0 0 0 1:1-0 0
H H 7. 0.2 M TEAB
6 FLO
6H 6H OH P, d-
o- \
OH
OH OH OH OH
19
C121121N2015P3
Exact Mass: 526.02 18
C121119N2014P3
Exact Mass: 508.00
1-Propyl-pseudo-UTP: A solution of 1-propyl-pseudouridine 16 (130.0 mg, 0.45
mmol; applied heat to
make it soluble) and proton sponge (144.66 mg, 0.67 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8
mL) was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (84.0 1_,
0.90 mmol, 2.0 equiv.)
was added dropwise to the solution and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (436.75 1_, 1.8 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(740.7 mg, 1.35 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 25.0 mL of water and the clear solution was stirred
vigorously for about
an hour at room temperature. The pH of the solution was adjusted to 6.50 by
adding about 3.5 mL of
1.0 M TEAB buffer along with vigorous stirring for about 3.0 hours. LCMS
analysis indicated the
formation of the corresponding triphosphate. The reaction mixture was then
lyophilized overnight. The
crude reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x
30.0 mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A =
100 mM TEAB buffer,
B = ACN; flow rate: 20.0 mL/min; retention time: 18.66-19.45 min). Fractions
containing the desired
were pooled and lyophilized to yield the 1-Propyl-pseudo-UTP as a
tetrakis(triethylammonium salt)
(63.33 mg, 26.66 %, based on C271 = 8,500). UVmax = 271 nm; MS: m/e 524.70 (M-
H).
- 244 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 18. Synthesis of 1-(2,2,2-trifluoroethyl)pseudo-UTP (03601015005
(175))
Scheme 8
0
0 0
FIN NH U
HN
NH /\CF3
HN
HO 0 Ac20, DMF 1 BSA
-30 C, DMA; Ac 0 2 TfOCH2CH3 __ Ac0 0
Hd OH
Acd 'OAc
1 Acd 0Ac
9
0
/-----CF3
HN N
NH3/Me0H HO
Hd OH
21
Synthesis of Compound 20: To a solution of 2',3',5'-tri-O-acetyl pseudouridine
9 (0.8 g, 2.2 mmol) in
dry CH3CN (20 mL) was added N,0-bis(trimethylsily1)acetamide (BSA) (3.0 mL),
and the reaction
5 mixture was reflux for 2 h. The reaction mixture was then cooled to room
temperature. To this
mixture, CF3CH20Tf (0.75 g, 3.3 mmol) was added, and the reaction mixture was
stirred at 60 C
overnight. More CF3CH20Tf (0.75 g, 3.3 mmol) was then added, and the reaction
mixture was stirred
at 60 C overnight. The reaction mixture was concentrated under reduced
pressure. The residue was
dissolved in CH2Cl2 (100 mL), washed with 1% NaHCO3 solution (3 x 50 mL),
dried (Na2504) and
10 evaporated to dryness. The residual was purified by silica gel column
using PE: EA (5:1 to 1:1) as the
eluent to give 0.7 g (72%) of product 20.
1-(2, 2, 2-Trifluoroethyl)pseudo-U (21): A solution of compound 20 (0.7 g) in
ammonia saturated
methanol (50 mL) was stirred at room temperature overnight. The volatiles were
removed under
15 reduced pressure. The residue was purified by silica gel column
chromatography, eluted with 5-10%
methanol in dichloromethane to give 260 mg compound 21 as pale yellow foam
with 98.66% HPLC
purity. 1H-NMR (DMSO-d6, 300 MHz, ppm) 6 11.62 (br, 1H), 7.79 (s, 1H), 5.01
(d, J =3.60Hz, 1H),
4.80 (d, J = 4.20Hz, 1H), 4.75 (t, J = 3.70Hz, 1H), 4.61 (q, J = 6.60Hz, 1H),
4.48 (d, J = 2.70Hz, 1H),
3.83-3.93 (m, 2H), 3.71 (d, J= 2.40Hz, 1H), 3.61-3.65 (m, 1H), 3.43-3.49 (m,
1H). The structure was
20 also verified by HMBC NMR.
- 245 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Scheme 9
HNN/F3 HNN/
C CF3
O 9
HO o 1. Trimethyl phosphate CI-P-0
2. Proton Sponge 4. Bu3N
OH OH OH OH
5. Bis(tributylammonium)
21 3. POCI3
22 pyrophosphate
C11H13F3N206
Exact Mass: 326.07 6. ACN
0 0
HNN/CF3 HNN/CF3
O OH
9 9 9 7Ø2 M TEAB
-r 0 P/-0
OH CH CH 213-0
0'
OH
OH OH OH OH
24
C11H16F3N2015P3 23
Exact Mass: 565.97
C11H14F3N2014P3
Exact Mass: 547.96
1-(2,2,2-Trifluoroethyl)pseudo-UTP: A solution of 1-(2,2,2-
trifluoroethyl)pseudouridine 21 (135.6 mg,
0.42 mmol; applied heat to make it soluble) and proton sponge (135.01 mg, 0.63
mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL) was stirred for 10.0 minutes at 0 C. Phosphorus
oxychloride (78.4.0 1_,
0.84 mmol, 2.0 equiv.) was added dropwise to the solution and it was then kept
stirring for 2.0 hours
under N2 atmosphere. A mixture of tributylamine (407.63 1_, 1.68 mmol, 4.0
equiv.) and
bis(tributylammonium) pyrophosphate (691.32 mg, 1.26 mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was
added at once. After -25 minutes, the reaction was quenched with 25.0 mL of
water, and the clear
solution was stirred vigorously for about an hour at room temperature. The pH
of the solution was
adjusted to 6.53 by adding about 3.6 mL of 1.0 M TEAB buffer along with
vigorous stirring for about
3.0 hours. LCMS analysis indicated the formation of the corresponding
triphosphate. The reaction
mixture was then lyophilized overnight. The crude reaction mixture was HPLC
purified (Shimadzu,
Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron; gradient (1 %):
100 % A for 3.0
min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0 mL/min;
retention time:
19.33-20.74 min). Fractions containing the desired were pooled and lyophilized
to yield the 1-(2,2,2-
Trifluoroethyl)pseudo-UTP as a tetrakis(triethylammonium salt) (93.88 mg,
39.52 %, based on C271 =
9,000). UVmax = 262 nm; MS: m/e 564.65 (M-H).
- 246 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Example 19. Synthesis of 2-thio-pseudo-UTP (00901015006 (193))
Scheme 10
0¨ 0
0
/
MeN NI
,--Me NH2
0¨ S __ K
HN NH dimethylformamide
NH
2
dimethyl acetal 0
HO 0 ______________ HO
o Et0Na
DMSO, heat Et0H
heat
H
HO OH O OH
1,3-dimethylpseudouridine
[64272-68-0] JJ
1 25 HN NH
0
H0c5)
HO OH
Compound 22
2-thiopseudouridine
26
Synthesis of N1,N3-Dimethylpseudouridine (25): A suspension of pseudouridine
(1) (1.0 g, 4.1 mmol)
in N,N-dimethylformamide dimethyl acetal (10 mL) was refluxed at 110 C for 1
h until a clear solution
was obtained. TLC (DCM-Me0H = 9:1) indicated the reaction was almost
completed. The solution
was concentrated in vacuo to give syrup which was triturated with a small
amount of methanol to give
640 mg solid product. The filtrate was concentrated and then further purified
by flash chromatography
on a silica gel column using DCM-Me0H 30:1 to 10:1 gradient eluent to give
additional 200 mg
product resulting in the total yield of 75.4%.
2-Thio-pseudo-U (26): A mixture of compound 25 (680 mg, 2.5 mmol) and thiourea
(950 mg, 12.5
mmol) in 1 M ethanolic sodium ethoxide (25 mL) was refluxed with stirring for
2 h. TLC (DCM-Me0H
= 9:1) indicated completion of the reaction. After cooling, 3M hydrochloric
acid was added to adjust
the pH to neutral, and the mercapto compound smell was noticed. It was then
adjusted to week basic
with ammonium hydroxide. It was purified by flash chromatography on a silica
gel column using DCM-
Me0H 20:1 to 10:1 to 5:1 gradient eluent giving 310 mg product in 47.7% yield.
This material
contained 69% beta-anomer and 28% alpha-anomer. It was then further purified
by preparative TLC
to give 230 mg pure beta-anomer product 26. The second preparative TLC
purification generated 183
mg final product with 94.23% HPLC purity. It was characterized by NMR and MS
spectral analysis.
- 247 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Scheme 11
HN A NH HN A NH
C) 0
I
HO- CI-P-0-
0
4. Bu3N
1. Trimethyl phosphate
2. Proton Sponge CI
OH OH OH OH 5. Bis(tributylammonium)
26 3. POCI3
27
pyrophosphate
C9H12N2055 6. ACN
Exact Mass: 260.05
HN A NH HN A NH
0, OH
0 0 0
H H 7Ø2 M TEAB
HO-P-O-P-O-P-0- o 0 P/-0
OH OH OH P, d-
o- \
OH
OH OH OH OH
29
C9H15N2014P35
Exact Mass: 499.95 28
C9H13N2013P35
Exact Mass: 481.94
2-Thio-pseudo-UTP: A solution of 2-Thiopseudouridine 26 (100.5 mg, 0.39 mmol;
applied heat to
make it soluble) and proton sponge (125.37 mg, 0.59 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8
mL) was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (72.8 pL,
0.78 mmol, 2.0 equiv.)
was added dropwise to the solution and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (378.52 pL, 1.56 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(641.94 mg, 1.17 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 25.0 mL of water, and the clear solution was
stirred vigorously for about
an hour at room temperature. The pH of the solution was adjusted to 6.75 by
adding about 3.5 mL of
1.0 M TEAB buffer along with vigorous stirring for about 3.0 hours. LCMS
analysis indicated the
formation of the corresponding triphosphate. The reaction mixture was then
lyophilized overnight. The
crude reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x
30.0 mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A =
100 mM TEAB buffer,
B = ACN; flow rate: 20.0 mL/min; retention time: 17.06-18.18 min). Fractions
containing the desired
were pooled and lyophilized to yield the 2-Thio-pseudo-UTP as a
tetrakis(triethylammonium salt)
(67.13 mg, 34.36%, based on a269 = 10,000). UVmax = 269 nm; MS: m/e 498.75 (M-
H).
- 248 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 20. Synthesis of 5-trifluoromethyl-UTP (00901013002 (352))
Scheme 12
0 00
F3C'-)LNH F3C'-)LNH
F3C'}'NH
NO 1. Trimethyl phosphate 0NO 0, pH
P p
N"
HO-1O 2. Proton Sponge CI-A- Os
P/-0
CI 4. Bu3N
3. POCI3 5. Bis(tributylammonium) H
OH OH OH OH OH OH
_ pyrophosphate
30 31 32
6. ACN
C10H11F3N206
C10H12P3N2014P3
Exact Mass: 312.06 Exact Mass:
533.95
F3C0
II 7Ø2 M TEAB
NH
I _L
0 0 0
OH OH OH
OH OH
33
C10H14F3N2015P3
Exact Mass: 551.96
5-Trifluoromethyl-UTP: A solution of 5-Trifluoromethyluridine 30(101 mg, 0.32
mmol; applied heat to
make it soluble) and proton sponge (102.86 mg, 0.48 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8
mL) was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (59.73 pL,
0.64 mmol, 2.0 equiv.)
was added dropwise to the solution and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (310.85 pL, 1.56 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(526.72 mg, 0.96 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 0.2 M TEAB buffer (13.7 mL) and the clear solution
was stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
time: 26.69-27.87 min). Fractions containing the desired were pooled and
lyophilized to yield the 5-
Trifluoromethyl-UTP as a tetrakis(triethylammonium salt) (34.11 mg, 19.30%,
based on a260 =
1 0 , 000) . UVmax = 258 nm; MS: m/e 550.65 (M-H).
- 249 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 21. Synthesis of 5-trifluoromethyl-CTP (00901014003 (351))
Scheme 13
NH2 NH2
NH2
F3C
F3C
F3C
I _L
0 1. Trimethyl phosphate
NO
HO-13 2. Proton Sponge 9
0, pH
Cl-p - _
0 -1:) 4. Bu3N µP -Os p
,13
CI -P-0
3. POCI3 0' \
5. Bis(trib 2-0-1
utylammonium) OH
OH OH OH OH OH OH
34 35 pyrophosphate
6. ACN 36
C10H12F3N305
C10H13F3N3013P3
Exact Mass: 311.07
Exact Mass: 532.96
NH2
F3C 7. 0.2 M TEAB
9 9 9
HO-p -0 -7 -0-7 -0-13
OH OH OH
OH OH
37
C10H15F3N3014P3
Exact Mass: 550.97
5-Trifluoromethyl-CTP: A solution of 5-Trifluoromethylcytidine 34 (109 mg,
0.35 mmol; applied heat to
make it soluble) and proton sponge (112.5 mg, 0.52 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL)
was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (65.34 pL, 0.70
mmol, 2.0 equiv.) was
added dropwise to the solution and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (340.00 pL, 1.40 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(576.10 mg, 1.05 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 0.2 M TEAB buffer (16.5 mL) and the clear solution
was stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
time: 17.77-18.63 min). Fractions containing the desired were pooled and
lyophilized to yield the 5-
Trifluoromethyl-CTP as a tetrakis(triethylammonium salt) (50.75 mg, 26.28 %,
based on a269 = 9,000).
UVmax = 269 nm; MS: m/e 549.65 (M-H).
- 250 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 22. Synthesis of 3-methyl-pseudo-UTP (00901015187 (236))
Scheme 14
0 0 0
A ,CH3 A ,CH3
,CH3
HN N HN N
HN N
0, pH
(:) Trimethyl phosphate
HO-1O 2. Proton Sponge 9
Cl-p-O 4. Bu3N =P -Os p
CI P-0
3. POCI3 \
5. Bis(tributylammonium) OH
OH OH OH OH
OH OH
38 39 _ pyrophosphate
6. ACN 40
C10H14N206
C10H15N2014P3
Exact Mass: 258.09
Exact Mass: 479.97
0
A CH3
HN 7. 0.2 M
TEAB
9 9 9
OH OH OH
OH OH
41
C10H17N2015123
Exact Mass: 497.98
3-Methyl-pseudo-UTP: A solution of 3-Methylpseudouridine 38 (104 mg, 0.4 mmol;
applied heat to
make it soluble) and proton sponge (128.58 mg, 0.6 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL)
was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (74.70 pL, 0.80
mmol, 2.0 equiv.) was
added dropwise to the solution, and it was then kept stirring for 2.0 hours
under N2 atmosphere. A
mixture of tributylamine (388.56 pL, 1.60 mmol, 4.0 equiv.) and
bis(tributylammonium) pyrophosphate
(658.40 mg, 1.05 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once.
After -25 minutes, the
reaction was quenched with 0.2 M TEAB buffer (17.0 mL) and the clear solution
was stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
time: 15.61-17.21 min). Fractions containing the desired were pooled and
lyophilized to yield the 3-
Methyl-pseudo-UTP as a tetrakis-(triethylammonium salt) (52.38 mg, 26.25 %,
based on a264 =
8,000). UVmax = 264 nm; MS: m/e 496.75 (M-H).
- 251 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 23. Synthesis of 5-methyl-2-thio-UTP (00901013003 (4))
Scheme 15
0 0 0
H3Cjt, H3Cjt,
NH NH
NH
I ,L
,
-"'S 1. Trimethyl phosphate 0NS 00H
.12/-0s 0 NS
HO OH OH 2. O
Sponge CI-CAI- OH OH _ 4p..y BrophNo
ibsupthyalatem
monium)
42
0\ H
OH OH
4344
6. ACN
C101-102056
CioHi5N2013P3S
Exact Mass: 274.06
Exact Mass: 495.95
0
7. 02 M
H3Cjt,
NH TEAB
0 0 0
OH CH CH
OH OH
C10H17N2014P3S
Exact Mass: 513.96
5-Methyl-2-thio-UTP: A solution of 5-Methyl-2-thiouridine 42 (55 mg, 0.2 mmol;
applied heat to make
5 it soluble) and proton sponge (64.30 mg, 0.3 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL) was
stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (37.35 pL, 0.40 mmol,
2.0 equiv.) was added
dropwise to the solution and it was then kept stirring for 2.0 hours under N2
atmosphere. A mixture of
tributylamine (194.28 pL, 0.8 mmol, 4.0 equiv.), and bis(tributylammonium)
pyrophosphate (329.20
mg, 0.6 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once. After -
25 minutes, the reaction
10 was quenched with 0.2 M TEAB buffer (8.5 mL) and the clear solution was
stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
15 time: 18.21-18.92 min). Fractions containing the desired were pooled and
lyophilized to yield the 5-
Methy1-2-thio-UTP as a tetrakis(triethylammonium salt) (62.44 mg, 60.00 %,
based on a276 = 13,120).
UVmax = 276 nm; MS: m/e 512.70 (M-H).
- 252 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 24. Synthesis of N4-methyl-CTP (00901014004 (346))
Scheme 16
NHCH3 NHCH3
NHCH3
NO
1. Trimethyl phosphate 0NO 0, pH
0 N
2. Proton Sponge CI-A- o's
CI 4. Bu3N
_________________________ 0 __________________________ 0 -P-Oz
3. POCI3
_ 5. Bis(tributylammonium) OOH
OH OH OH OH OH OH
pyrophosphate
46 47 48
C10H15N305 6. ACN
Exact Mass: 257.10
C10H16143013P3
Exact Mass: 478.99
NHCH3 7. 0.2 M
TEAB
I _L
0 0 0
H04-04-04-0-13
OH OH OH
OH OH
49
C10H18143014P3
Exact Mass: 497.00
N4-Methyl-CTP: A solution of N4-Methyl-cytidine 46 (100.7 mg, 0.39 mmol;
applied heat to make it
soluble) and proton sponge (126.44 mg, 0.59 mmol, 1.5 equiv.) in trimethyl
phosphate (0.8 mL) was
stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (72.8 pL, 0.78 mmol,
2.0 equiv.) was added
dropwise to the solution, and it was then kept stirring for 2.0 hours under N2
atmosphere. A mixture of
tributylamine (378.85 pL, 1.56 mmol, 4.0 equiv.) and bis(tributylammonium)
pyrophosphate (642.0
mg, 1.17 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once. After -
25 minutes, the reaction
was quenched with 0.2 M TEAB buffer (17.0 mL) and the clear solution was
stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
time: 17.05-17.80 min). Fractions containing the desired were pooled and
lyophilized to yield the N4-
Methyl-CTP as a tetrakis(triethylammonium salt) (35.05 mg, 17.94 %, based on
a270 = 11,000).
UVmax = 270 nm; MS: m/e 495.70 (M-H).
- 253 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 25. Synthesis of 5-hydroxymethyl-CTP (00901014005 (350))
Scheme 17
NH2 NH2
NH2
TBSOL- N TBSON
HON
NO I L
I _L
1. Trimethyl phosphate 0 _ 0, pH
9
NO
2. Proton Sponge 0 P-0
HO 4. Bu3N
CI -P-0
3. POCI3 0' \
5. Bis(tributylammonium) OH
OH OH OH OH OH OH
_ pyrophosphate
50 5152
6. ACN
C16H2914306S
C10H16N3014P3
,
Exact Mass: 387.18
Exact Mass: 494.98
Ho NH2 7T E0A113 M
0 0 0
OH OH OH
OH OH
53
C10H18N3015P3
Exact Mass: 513.00
5-Hydroxymethyl-CTP: A solution of 5-0TBS-CH2-cytidine 50 (126.0 mg, 0.33
mmol; applied heat to
make it soluble) and proton sponge (107.2 mg, 0.5 mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL)
was stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (61.6 pL, 0.66
mmol, 2.0 equiv.) was
added dropwise to the solution, and it was then kept stirring for 2.0 hours
under N2 atmosphere. The
TBS group had been removed during POCI3 reaction and corresponding
monophosphate (without
TBS) was detected by LCMS. A mixture of tributylamine (320.28 pL, 1.32 mmol,
4.0 equiv.) and
bis(tributylammonium) pyrophosphate (543.2 mg, 0.99 mmol, 3.0 equiv.) in
acetonitrile (2.3 mL) was
added at once. After -25 minutes, the reaction was quenched with 0.2 M TEAB
buffer (13.0 mL) and
the clear solution was stirred at room temperature for an hour. LCMS analysis
indicated the formation
of corresponding triphosphate (without TBS). The reaction mixture was then
lyophilized overnight.
The crude reaction mixture was HPLC purified (Shimadzu, Phenomenex C18
preparative column,
250 x 30.0 mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min,
A = 100 mM TEAB
buffer, B = ACN; flow rate: 20.0 mL/min; retention time: 16.48-17.36 min).
Fractions containing the
desired were pooled and lyophilized to yield the 5-Hydroxymethyl-CTP as a
tetrakis(triethylammonium salt) (16.72 mg, 9.75% for two steps, based on a276
= 9,000). UVmax =
276 nm; MS: m/e 511.70 (M-H).
- 254 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 26. Synthesis of 3-methyl-CTP (00901014006)
Scheme 18
NH NH NH
,CH3 ,CH3
NO
,CH3
1. Trimethyl phosphate 0NO 0, pH
0 NO
HO- () 2. Proton Sponge 4. Bu3N
Ol -P-0
3. POCI3 0' \
5. Bis(tributylammonium) OH
OH OH OH OH OH OH
54
_ pyrophosphate
6. ACN 56
CioHi5N305
C10H16N3013P3
Exact Mass: 257.10
Exact Mass: 478.99
NH 7. 0.2 M
AN,CH3 TEAB
0 0 0
HO-P-04-04-0-3
OH OH OH
OH OH
57
C10H18N3014P3
Exact Mass: 497.00
3-Methyl-CTP: A solution of 3-Methyl-cytidine 54 (93.0 mg, 0.36 mmol; applied
heat to make it
5 soluble) and proton sponge (115.7 mg, 0.54 mmol, 1.5 equiv.) in trimethyl
phosphate (0.8 mL) was
stirred for 10.0 minutes at 0 C. Phosphorus oxychloride (67.2 pL, 0.72 mmol,
2.0 equiv.) was added
dropwise to the solution, and it was then kept stirring for 2.0 hours under N2
atmosphere. A mixture of
tributylamine (349.4 pL, 1.44 mmol, 4.0 equiv.) and bis(tributylammonium)
pyrophosphate (592.6 mg,
1.08 mmol, 3.0 equiv.) in acetonitrile (2.5 mL) was added at once. After -25
minutes, the reaction
10 was quenched with 0.2 M TEAB buffer (17.0 mL), and the clear solution
was stirred at room
temperature for an hour. LCMS analysis indicated the formation of the
corresponding triphosphate.
The reaction mixture was then lyophilized overnight. The crude reaction
mixture was HPLC purified
(Shimadzu, Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron;
gradient (1 %): 100 %
A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0
mL/min; retention
15 time: 16.15-16.67 min). Fractions containing the desired were pooled and
lyophilized to yield the 3-
Methyl-CTP as a tetrakis(triethylammonium salt) (20.4 mg, 11.4 %, based on
a277 = 9,000). UVmax =
277 nm; MS: m/e 495.75 (M-H).
- 255 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 27. Synthesis of UTP-oxyacetic acid Me ester (00901013004) (348))
Scheme 19
0 0 0 0
o-
HNõ.0 e Me
CD-1\1
0 N 1. Trimethyl phosphate
9 0 N
HO 2. Proton Sponge CI-P-0
3. POCI3 CI 4. Bu3N
OH OH OH OH 5. Bis(tributylammonium)
58 59 pyrophosphate
Ci2Hi6N203
6. ACN
Exact Mass: 332.09
0 0
HN 0 0 -
.K.,...õØõ,o-Me
o-Me
0 N 0, pH 0 N
9 9 9 7. 0.2 M TEAB
ci PO
OH OH OH
0' \
OH
OH OH OH OH
61
C1211131µ12018P3
Exact Mass: 571.98
C12H17N2017P3
Exact Mass: 553.97
UTP-5-oxyacetic acid Me ester: A solution of Uridine-5-oxyacetic acid Me ester
58 (100.3 mg, 0.3
5 mmol; applied heat to make it soluble) and proton sponge (96.44 mg, 0.45
mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL) was stirred for 10.0 minutes at 0 C. Phosphorus
oxychloride (56.0 pL,
0.6 mmol, 2.0 equiv.) was added dropwise to the solution and it was then kept
stirring for 2.0 hours
under N2 atmosphere. A mixture of tributylamine (291.2 pL, 1.2 mmol, 4.0
equiv.) and
bis(tributylammonium) pyrophosphate (493.8 mg, 0.9 mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was
10 added at once. After -25 minutes, the reaction was quenched with 0.2 M
TEAB buffer (14.2 mL) and
the clear solution was stirred at room temperature for an hour. LCMS analysis
indicated the formation
of the corresponding triphosphate. The reaction mixture was then lyophilized
overnight. The crude
reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x 30.0
mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A = 100 mM
TEAB buffer, B =
15 ACN; flow rate: 20.0 mL/min; retention time: 18.52-19.06 min). Fractions
containing the desired were
pooled and lyophilized to yield the UTP-5-oxyacetic acid Me ester as a
tetrakis(triethylammonium
salt) (20.04 mg, 11.67 `)/0, based on a275 = 10,000). UVmax = 275 nm; MS: m/e
570.65 (M-H).
- 256 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 28. Synthesis of 5-methoxycarbonylmethyl-UTP (00901013005 (358))
Scheme 20
0 0
J-
J-1,0CH3 r
HN , OCH3 HN
O'Ni 0 C/N 0
1. Trimethyl phosphate 9
HO7 ()
2. Proton Sponge
4. Bu3N
OH OH 3. POCI3 OH OH 5. Bis(tributylammonium)
62 63
pyrophosphate
C12H16N208 6. ACN
Exact Mass: 316.09
0
HN 0
)rOCH3
OCH3
HN
0 jHin
0 N ON 0, pH
9 9 9 7Ø2 M TEAB 'µP-0,
Os /13-0
OH OH OH -P-0
0'
OH
OH OH OH OH
C12H19N2017133 64
Exact Mass: 555.99
C12H17N2016P3
Exact Mass: 537.98
5-Methoxycarbonylmethyl-UTP: A solution of 5-Methoxycarbonylmethyl-uridine 62
(101.0 mg, 0.32
5 mmol; applied heat to make it soluble) and proton sponge (102.86 mg, 0.48
mmol, 1.5 equiv.) in
trimethyl phosphate (0.8 mL) was stirred for 10.0 minutes at 0 C. Phosphorus
oxychloride (59.73 pL,
0.64 mmol, 2.0 equiv.) was added dropwise to the solution and it was then kept
stirring for 2.0 hours
under N2 atmosphere. A mixture of tributylamine (310.58 pL, 1.28 mmol, 4.0
equiv.) and
bis(tributylammonium) pyrophosphate (526.72 mg, 0.9 mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was
10 added at once. After -25 minutes, the reaction was quenched with 0.2 M
TEAB buffer (15.1 mL) and
the clear solution was stirred at room temperature for an hour. LCMS analysis
indicated the formation
of the corresponding triphosphate. The reaction mixture was then lyophilized
overnight. The crude
reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x 30.0
mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A = 100 mM
TEAB buffer, B =
15 ACN; flow rate: 20.0 mL/min; retention time: 17.15-18.38 min). Fractions
containing the desired were
pooled and lyophilized to yield the 5-Methoxycarbonylmethyl-UTP as a
tetrakis(triethylammonium
salt) (49.88 mg, 28.12 %, based on a265 = 11,000). UVmax = 265 nm; MS: m/e
554.70 (M-H).
- 257 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 29. Synthesis of 5-methylaminomethyl-UTP (00901013006 (353))
Scheme 21
0 0
HN)-N-CH3
HNN,CH3
0 N 0 CF3 o 0 N 0 CF3
CI-P-0
1. Trimethyl phosphate 61
4. Bu3N
2. Proton Sponge
OH OH OH OH 5.
Bis(tributylammonium)
66 3. POCI3
67
pyrophosphate
6. ACN
C13H16F3N307
Exact Mass:
383.09
0 0
HN HN
CH3
CH3
0 N 0 CF3 )-
)-
0, OH
ON 0 CF3
0 0 0 p
H 7Ø2 M TEAB
P-C)
OH OH OH 213-0
0'
OH
OH OH OH OH
69
C131-119F3N3016P3
Exact Mass: 622.99
68
C131-117F3N3015P3
Exact Mass: 604.98
Scheme 22
0 0
HN N
-CH3 HN
CH3
,
ON OCF3 H
0 0 0 0 0 0
OH
HO-P-O-P-O-P-0- NH4
0 HO-P-O-
P-O-P-0- 0
OH OH OH Overnight
OH OH OH
OH OH OH OH
69 70
CtilizoN30.15P3
Ci3H.19F3N30.16P3
Exact Mass: 622.99 Exact Mass:
527.01
5-Methylaminomethyl-UTP: A solution of 5-N-TFA-N-Methylaminomethyl-uridine 66
(110.0 mg, 0.29
mmol; applied heat to make it soluble) and proton sponge (94.30 mg, 0.44 mmol,
1.5 equiv.) in
trimethyl phosphate (0.8 mL) was stirred for 10.0 minutes at 0 C. Phosphorus
oxychloride (54.13 pL,
0.58 mmol, 2.0 equiv.) was added dropwise to the solution and it was then kept
stirring for 2.0 hours
under N2 atmosphere. A mixture of tributylamine (281.46 pL, 1.16 mmol, 4.0
equiv.) and
bis(tributylammonium) pyrophosphate (477.34 mg, 0.87 mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was
added at once. After -25 minutes, the reaction was quenched with 0.2 M TEAB
buffer (13.7 mL) and
the clear solution was stirred at room temperature for an hour. LCMS analysis
indicated the formation
of the corresponding triphosphate. To this above crude reaction mixture, about
22.0 mL of
concentrated NH4OH was added and the reaction mixture was stirred at room
temperature overnight.
It was then lyophilized overnight and the crude reaction mixture was HPLC
purified (Shimadzu,
Phenomenex C18 preparative column, 250 x 30.0 mm, 5.0 micron; gradient (1 %):
100 % A for 3.0
min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0 mL/min;
retention time:
- 258 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
14.89-16.11 min). Fractions containing the desired were pooled and lyophilized
to yield the 5-
Methylaminomethyl-UTP as a tetrakis(triethylammonium salt) (35.27 mg, 19.31 %
for two steps,
based on a266 = 10,000). UVmax = 266 nm; MS: m/e 525.70 (M-H).
Example 30. Synthesis of N4,N4,2'-0-trimethyl-CTP (03601074029)
Scheme 23
H3C, ,CH3 H3C, ,CH3
1. Trimethyl phosphate
9
HO 2. Proton Sponge CI-P-0
3. POCI3 CI 4. Bu3N
OH OCH3 OH OCH3 5.
Bis(tributylammonium)
71 72 pyrophosphate
O12H131\1303 6. ACN
Exact Mass: 285.13
H3C, ,CH3 H3C, ,CH3
NO c) OH NO
9 9 9 7. 0.2 M TEAB
-40 tp-0-1,
OH OH OH -P-0
0' \
OH
OH OCH3 OH OCH3
74
C12H22N3014P3 73
Exact Mass: 525.03
C12H20N3013P3
Exact Mass: 507.02
N4, N4, 2'-0-Trimethyl-CTP (74): A solution of N4, N4, 2'-0-trimethyl-cytidine
71 (101.5 mg, 0.36
mmol; applied heat to make it soluble) and proton sponge (115.7 mg, 0.54 mmol,
1.5 equiv.) in
trimethyl phosphate (0.8 mL) was stirred for 10.0 minutes at 0 C. Phosphorus
oxychloride (67.20 pL,
0.72 mmol, 2.0 equiv.) was added dropwise to the solution and it was then kept
stirring for 2.0 hours
under N2 atmosphere. A mixture of tributylamine (349.40 pL, 1.44 mmol, 4.0
equiv.) and
bis(tributylammonium) pyrophosphate (592.60 mg, 1.08 mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was
added at once. After -25 minutes, the reaction was quenched with 0.2 M TEAB
buffer (17.0 mL) and
the clear solution was stirred at room temperature for an hour. LCMS analysis
indicated the formation
of the corresponding triphosphate. The reaction mixture was then lyophilized
overnight. The crude
reaction mixture was HPLC purified (Shimadzu, Phenomenex C18 preparative
column, 250 x 30.0
mm, 5.0 micron; gradient (1 %): 100 % A for 3.0 min, then 1% B/min, A = 100 mM
TEAB buffer, B =
ACN; flow rate: 20.0 mL/min; retention time: 18.67-19.38 min). Fractions
containing the desired were
pooled and lyophilized to yield the N4, N4, 2'-0-Trimethyl-CTP (74) as a
tetrakis(triethylammonium
salt) (30.22 mg, 16.11 `)/0, based on a278 = 9,000). UVmax = 278 nm; MS: m/e
523.75 (M-H).
- 259 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 31. Synthesis of 5-methoxycarbonylmethy1-2'-0-methyl-UTP (00901073005
(78))
Scheme 24
0 0
OCH3
HN
0
0
1. Trimethyl phosphate 9
2. Proton Sponge CI-P-
61
__________________________ > 4. Bu3N
OH OCH3 3. POCI3 OH OCH3 5.
Bis(tributylammonium)
75 76 pyrophosphate
C13H18N208 6. ACN
Exact Mass: 330.11
00
OCH3 )I,,.õ,ThrOCH3
HNJf
HN
0 N 0
0, pH
9 9 9 7. 0.2 M TEAB =P -Os p
P -0
0-
-[2:L
OH OH OH .212-6
OH
OH OCH3 OH OCH3
78
77
C13H21N2017123
Exact Mass: 570.01
C13H19N2016123
Exact Mass: 551.99
5-Methoxycarbonylmethy1-2'-0-methyl-UTP (78): A solution of 5-
Methoxycarbonylmethy1-2'-0-methyl-
uridine 75 (102.0 mg, 0.31 mmol; applied heat to make it soluble) and proton
sponge (100.72 mg,
0.47 mmol, 1.5 equiv.) in trimethyl phosphate (0.8 mL) was stirred for 10.0
minutes at 0 C.
Phosphorus oxychloride (57.87 pL, 0.62 mmol, 2.0 equiv.) was added dropwise to
the solution and it
was then kept stirring for 2.0 hours under N2 atmosphere. A mixture of
tributylamine (300.87 pL, 1.24
mmol, 4.0 equiv.) and bis(tributylammonium) pyrophosphate (510.26 mg, 0.93
mmol, 3.0 equiv.) in
acetonitrile (2.5 mL) was added at once. After -25 minutes, the reaction was
quenched with 0.2 M
TEAB buffer (14.64 mL) and the clear solution was stirred at room temperature
for an hour. LCMS
analysis indicated the formation of the corresponding triphosphate. The
reaction mixture was then
lyophilized overnight. The crude reaction mixture was HPLC purified (Shimadzu,
Phenomenex C18
preparative column, 250 x 30.0 mm, 5.0 micron; gradient (1 %): 100 % A for 3.0
min, then 1% B/min,
A = 100 mM TEAB buffer, B = ACN; flow rate: 20.0 mL/min; retention time: 18.57-
19.35 min).
Fractions containing the desired were pooled and lyophilized to yield the 5-
Methoxycarbonylmethy1-
2'-0-methyl-UTP (78) as a tetrakis(triethylammonium salt) (54.60 mg, 30.97 %,
based on a265 =
11,000). UVmax = 265 nm; MS: m/e 568.65 (M-H).
- 260 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 32. Synthesis of 5-methoxy uridine (compound 15) and 5-methoxy UTP
(NTP of said
compound)
Scheme 25
0 0 0
H3cojL
H3co H3co
NH
NH
0 N 0 4. Bu3N 9 9 9
1. Trimethyl phosphate
HO-1_04 2. Proton Sponge 01 5. Bis(tributylammonium)
u.1 u
. .1 u
. .1.
H H H
pyrophosphate
OH OH 3. POC13 OH OH OH
OH
6Ø2 M TEAB buffer
153
16
0 10 207
CioHi7N2016123
4
Exact Mass: 513.98
Exact Mass: 274.08
A solution of 5-methoxy uridine (compound 15) (69.0 mg, 0.25 mmol, plus heat
to make it
soluble) was added to proton sponge (80.36 mg, 0.375 mmol, 1.50 equiv.) in 0.7
mL
trimethylphosphate (TMP) and was stirred for 10 minutes at 0 C. Phosphorous
oxychloride (POC13)
(46.7 ul, 0.50 mmol, 2.0 equiv.) was added dropwise to the solution before
being kept stirring for 2
hours under N2 atmosphere. After 2 hours the solution was reacted with a
mixture of
bistributylammonium pyrophosphate (TBAPP or (n-Bu3NH)2H2P207) (894.60 mg, 1.63
mmol, 6.50
equiv.) and tributylamine (243.0 ul, 1.00 mmol, 4.0 equiv.) in 2.0 ml of
dimethylformamide. After
approximately 15 minutes, the reaction was quenched with 17.0 ml of 0.2M
triethylammonium
bicarbonate (TEAB) and the clear solution was stirred at room temperature for
an hour. The reaction
mixture was lyophilized overnight and the crude reaction mixture was purified
by HPLC (Shimadzu,
Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron;
gradient: 100 % A
for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 10.0
mL/min; retention
time: 16.57-17.51 min). Fractions containing the desired compound were pooled
and lyophilized to
produce the NTP of compound 15. The triphosphorylation reactions were carried
out in a two-neck
flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were
dried over P205
under vacuum overnight prior to use. The formation of monophosphates was
monitored by LCMS.
- 261 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 33. Synthesis of 6-Methylpseudouridine (03601015037):Scheme 26
Cat. Na0Me TBDMS-CI Et3N
EtSH + CH20 ____________________________ EtSCH2OH ________ EtSCH20 Si
DMAP DCM
SO2Cl2
DCM
0
0
H
HN NH NNH
L(:) TBDMS-CI CICH20 Si __
HO TBDMSO
imidazole
BSA CH3CN
HO OH TBDMSO OTBDMS
1 79
0 0
N 0 HN N 0
-78 degree C
LDA, Mel 0 CH3
TBDMSO TBDMSO
TBDMSO OTBDMS TBDMSO OTBDMS
80 81
0
HN NH
TBAF
______________________ H(:) 0 CH3
HO OH
82
Ethylthiomethanol: To a stirred mixture of ethanethiol (7.4 ml, 6.2 g, 0.1
mol) and paraformaldehyde
(3.0 g, 0.1 mol) was added 0.03 mL saturated sodium methoxide solution in
methanol as catalyst. It
was stirred at 40 C for 30 min, and cooled to give liquid product 9.2 g. It
was used for next step
without further purification.
(tert-Butyldimethylsilyloxy)methyl ethyl sulfide: To a solution of
ethylthiomethanol (4.6 g, 50 mmol) in
50 mL of anhydrous dichloromethane was added tert-butyldimethylsilylchloride
(8.31 g, 55 mmol), 4-
(N,N-dimethylamino)pyridine (244 mg, 2 mmol) and triethylamine (8.35 ml, 60
mmol). The mixture
was stirred at ambient temperature under nitrogen atmosphere for 4 h, and
diluted with
dichloromethane. The mixture was washed successively with water (x2) and
saturated aqueous
ammonium chloride (x2), and then dried over anhydrous sodium sulfate. The
filtrate solution was
concentrated under reduced pressure to give 8.72 g product as pale yellow oil
in 84% yield. It was
used in next step without further purification.
- 262 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
(tert-Butyldimethylsilyloxy)methyl Chloride: A solution of (tert-
butyldimethylsilyloxy)methyl ethyl sulfide
(5.1 mg, 25 mmol) in anhydrous dichloromethane was cooled to 0 C. Su!fury
chloride (1.6 mL, 10
mmol) in 20 mL of anhydrous methylene chloride was added under stirring over
30 min. The reaction
mixture was stirred at room temperature for an additional 10 min, and
concentrated under reduced
pressure giving 4.2 g product as pale yellow oil, which was used directly for
next step without further
purification.
Compound 79: A mixture of pseudouridine (1) (3.0 g, 12.3 mmol), imidazole (4.2
g, 61.5 mmol, 5.0
eq), and t-butyldimethylsilyl chloride (7.4 g, 49.2 mmol, 4.0 eq) in anhydrous
DMF was stirred at 30
C overnight. TLC (PE-EA = 2:1) indicated completion of the reaction. The
reaction mixture was
treated with dichloromethane and saturated sodium carbonate solution. The
organic phase was
separated, and the aqueous phase was extracted with ethyl acetate. The
combined organic phase
was dried over anhydrous sodium sulfate. The filtrate was concentrated under
reduced pressure. The
crude product was purified by flash chromatography on a silica gel column
using PE-EA (3:1) as
eluent giving white foam product 79 which was used for next step without
further purification and
characterization.
Compound 80: A stirred mixture of trisilylated compound 79 (1.5 g, 2.56 mmol)
in 20 mL of anhydrous
acetonitrile and 8 mL of BSA was heated to 65 C under nitrogen atmosphere for
6 h. t-
(Butyldimethylsilyloxy)methyl chloride (1.8 g, 10 mmol) was added, and the
resulting reaction mixture
was stirred at 65 C overnight. TLC (PE-EA = 3:1) indicated completion of the
reaction. The reaction
mixture was cooled to room temperature and treated with dichloromethane and
aqueous saturated
sodium carbonate solution. The layers were separated, and the aqueous layer
was extracted with
dichloromethane (30 mL x3). The combined organic phase was dried over
anhydrous sodium sulfate,
and filtered. The solvent was evaporated under reduced pressure. The residue
was purified by flash
chromatography on a silica gel column giving 1.2 g desired product 80 in 64%
yield.
Compound 81: N,N-Diisopropylamine (1.4 mL, 10 mmol) was dissolved in 20 mL of
anhydrous THF.
The solution was cooled to -78 C under nitrogen atmosphere. n-Butyl lithium
(4 mL, 10 mmol; 2.5 M
in hexane) was added dropwise under stirring over 1 h. A solution of compound
80 (2.2 g, 3 mmol) in
5 mL of anhydrous THF was added to the LDA solution prepared above. The
resulting reaction
mixture was stirred at -78 C for an additional 2 h. During this time, a
solution of iodomthane (1.25
mL, 20 mmol) in 10 mL of anhydrous THF was cooled to -78 C under nitrogen
atmosphere. The LDA
solution of compound D at low temperature was directly transferred to this
cooled iodomethane
solution. The resulting reaction mixture was stirred at -78 C for 30 min. The
reaction mixture was
treated with aqueous ammonium chloride solution, and it was allowed to warm to
room temperature,
followed by the treatment with ethyl acetate and aqueous sodium bicarbonate
solution. The layers
were separated, and the aqueous phase was extracted with ethyl acetate. The
combined organic
phase was tried over anhydrous sodium sulfate and filtered. The solution was
concentrated under
reduced pressure. The residue was purified by flash chromatography on a silica
gel column providing
1.1 g desired 6-methylated product 82 in 49% yield.
- 263 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
6-Methylpseudouridine (82): Compound 81(1.1 g, 1.48 mmol) was treated with 0.5
M TBAF solution
in THF, and it was stirred at 30 C overnight. TLC indicated completion of the
reaction. The mixture
was concentrated and purified by flash chromatography on a silica gel column
providing 257 mg
desired product in 67% yield with 99.42% HPLC purity. It was characterized by
NMR and MS spectral
analysis.
Example 34. Synthesis of 6, N1-dimethylpseudouridine (03601015107):
Scheme 27
0
0
H
HNANH N ANH
TBDMS-CI
HO 0 ____________ TBDMSO BSA DCM
CH3I
imidazole
HO OH TBDMSO OTBDMS
1 79
0 0
HNAN/ HNAN/
-78 degree C
TBDMSO
0 LDA, Mel __ TBDMSO %J 0 r-13 TBAF
TBDMSO OTBDMS TBDMSO OTBDMS 0
83 84
HN AN
0 vr-13
HO
HO OH
10 Compound 83: Compound 79 (5.87 g, 10 mmol) was dissolved in 100 mL of
anhydrous
dichloromethane, and 20 mL of BSA was added. The mixture was refluxed under
nitrogen
atmosphere for 4 h. iodomethane (2.56 g, 1.12 mL, 1.8 eq) was added, and the
reaction mixture was
continued to be heated at reflux temperature for 5 days. TLC (PE-EA = 3:1)
indicated trace starting
material left. The reaction mixture was cooled to room temperature, and
treated with dichloromethane
15 and aqueous sodium bicarbonate solution. The layers were separated, and
the aqueous phase was
extracted with dichloromethane. The combined organic phase was dried over
anhydrous sodium
sulfate, and the filtrate was concentrated under reduced pressure. The residue
was purified by flash
chromatography on a silica gel column giving 3.9 g compound 83 as white foam
in 65% yield. Some
starting material was recovered.
- 264 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Compound 84: N,N-Diisopropylamine (1.4 mL, 10 mmol) was dissolved in 20 mL of
anhydrous THF.
The solution was cooled to -78 C under nitrogen atmosphere. n-Butyl lithium
(4 mL, 10 mmol; 2.5 M
in hexane) was added dropwise under stirring over 1 h. A solution of compound
83 (1.8 g, 3 mmol) in
mL of anhydrous THF was added to the LDA solution prepared above. The
resulting reaction
5 mixture was stirred at -78 C for an additional 2 h. During this time, a
solution of iodomthane (1.25
mL, 20 mmol) in 10 mL of anhydrous THF was cooled to -78 C under nitrogen
atmosphere. The LDA
solution of compound 83 at low temperature was directly transferred to this
cooled iodomethane
solution. The resulting reaction mixture was stirred at -78 C for 30 min. The
reaction mixture was
treated with aqueous ammonium chloride solution, and it was allowed to warm to
room temperature,
followed by the treatment with ethyl acetate and aqueous sodium bicarbonate
solution. The layers
were separated, and the aqueous phase was extracted with ethyl acetate. The
combined organic
phase was tried over anhydrous sodium sulfate and filtered. The solution was
concentrated under
reduced pressure. The residue was purified by flash chromatography on a silica
gel column providing
1.2 g desired product 84 as pale yellow foam in 65% yield.
1,6-Dimethylpseudouridine (85): Compound 84 (1.2 g, 1.95 mmol) was treated
with 10 mL of 1 M
TBAF solution in THF, and it was stirred at room temperature for 24 h. TLC
indicated completion of
the reaction. The mixture was concentrated and purified by flash
chromatography on a silica gel
column using methylene chloride-methanol (20:1) providing 240 mg desired
product 85 with 99.61%
HPLC purity. It was characterized by NMR and MS spectral analysis (see
separate document for
spectra).
Example 35. Synthesis of NI-Allylpseudouridine (03601015151):
Scheme 28
HNANH HNANH
HO TBDMS-CI
BSA CH3CN
imidazole TBDMSOcõ¨
HO OH r-r Br
1 TBDMSO OTBDMS 3-5
days
79
0 0
HN)-N /\
HNAN
C) TBAF _____ HO
TBDMSO
1¨r 1¨r
TBDMSO OTBDMS HO OH
86 87
- 265 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Compound 86: A stirred mixture of compound 79 (1.17g, 2.0 mmol) in 20 mL of
anhydrous
acetonitrile and 10 mL of BSA was heated to 65 C under nitrogen atmosphere
for 4 h. Ally! bromide
(0.5 mL, 0.7 g, 5.8 mmol) was added. The reaction mixture was stirred at 65 C
for an additional 24 h.
TLC (PE-EA = 3:1) indicated completion of the reaction. The cooled reaction
mixture was treated with
ethyl acetate and saturated sodium carbonate solution. The layers were
separated, and the aqueous
layer was extracted with ethyl acetate. The combined organic phase was dried
over anhydrous
sodium sulfate. The filtrate was concentrated under reduced pressure. The
residue was purified by
flash chromatography on a silica gel column using PE-EA as eluent giving 650
mg product 86 in 52%
yield (some starting material was recovered).
1-Allyl-pseudouridine (87): Compound C (1.1 g, 1.75 mmol) was dissolved in 10
mL of THF, and 10
mL of 1M TBAF in THF was added. The reaction mixture was stirred at room
temperature for 24 h.
The solvent was concentrated, and the residue was purified by flash
chromatography on a silica gel
column giving 284 mg desired product 87 in 57% yield with 95.47% yield. It was
characterized by
NMR and MS spectral analysis (see different document for spectra).
Example 36. Synthesis of 1-Propargyl-pseudouridine (03601015153):
Scheme 29
0 0
HNANH
HNANH
0 1) BSA, 2.5 eq
HO 0 CH2CH2
Im,TBDMSCI TBDMSO
_______________________________________________________________________ )11,
DMF 2) refluxed
7d
HO OH
TBDMSO OTBDMS
1
79
0 0
HN AN/ ¨
HNAN
TBDMSO
0 TBAF/THF
0
)0 HO
TBDMSO OTBDMS
HO OH
88 89
Synthesis of compound 88: Bis-trimethylsilylacetamide (BSA, 10 ml) was added
to a stirred solution
of Compound 79 (1.5 g, 2.56 mmol) in DCM (20 mL). After stirring for four hour
at 40 degree C,
propargyl bromide (0.36 mL) was added to the solution, and the solution was
then heated at reflux
temperature for 24 h. The reaction mixture was concentrated to dryness under
reduced pressure. The
residue was purified via silica gel chromatography using petroleum ether (PE):
ethyl acetate (EA) =
20:1 ¨ 8:1 to give 1.1 g compound 88 as light yellow foam in 81% yield.
- 266 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
1-Propargyl-pseudouridine (89): To a solution of Compound 88 (2.2 g, 1 eq) in
THF was added TBAF
in THF (1 M, 2 mL), and the mixture was stirred overnight at 30 degree C. The
mixture was
concentrated under reduced pressure to dryness. The resulted crude product was
purified by silica
gel chromatography using Me0H-DCM = 1:50 ¨ 1:25 to give 0.325 g product 89 as
light pink solid in
32.7% yield. HPLC purity: 98.2%; 1H NMR (DMSO-d6): 6 11.4 (s, 1H), 7.81 (s,
1H), 4.97-4.99 (d, 1H,
J = 3.9Hz), 4.77-4.78 (m, 2H), 4.46-4.50 (m, 3H), 3.83-3.93 (m, 2H), 3.59-
3.71(m, 2H), 3.42-3.50 (m,
2H).
Example 37. Synthesis of 1-Cyclopropylmethylpseudouridine (03601015030):
Scheme 30
0 0
HNANH HNANH
1) BSA, 2.5 eq
HO
Im,TBDMSCI
TBDMSO CH2CH2
0
DMF 2) refluxed 7d
HO OH TBDMSO OTBDMS
1 79
0
0
HNN
HNAN
TBAF/THF 0
TBDMSO
TBDMSO OTBDMS HO OH
90 91
Synthesis of compound 90: Bis-trimethylsilylacetamide (BSA, 5 ml) was added to
a stirred solution of
Compound 79 (1.5 g, 2.6 mmol) in DCM (15 mL). After stirring for four hour at
40 degree C,
cyclopropylmethyl bromide (0.45 mL) was added to the solution, and the
solution was then heated at
reflux temperature for 5 days. The reaction mixture was concentrated to
dryness under reduced
pressure. The residue was purified via silica gel chromatography using PE: EA
= 20:1-8:1 to give 1.1
g product 90 as light yellow foam in 67% yield.
1-Cyclopropylmethylpseudouridine (91): To a solution of Compound 90 (1.2 g, 1
eq) in THF was
added TBAF in THF (1 M, 2 mL), and the mixture was stirred overnight. The
mixture was
concentrated to dryness under reduced pressure. The resulting crude product
was purified by silica
gel chromatography using Me0H : DCM = 1:50-1:25 to give 0.26 g product 91 as
white solid in
46.6% yield. HPLC purity: 97.6%;1H NMR (DM50-d6): 6 11.28 (s, 1H), 7.84 (s,
1H), 4.97-4.99 (d, 1H,
J = 3.6Hz), 4.82-4.85 (t, 1H, J = 4.5Hz), 4.75-4.76 (d, 1H, J = 4.5Hz), 4.46-
4.47 (d, 1H, J = 3Hz),
- 267 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
3.88-3.95 (m, 2H), 3.58-3.71 (m, 3H), 3.34-3.45 (m, 2H), 1.11 (1H), 0.45-0.48
(m, 2H), 0.32-0.35 (m,
2H).
Example 38. Synthesis of 6-Chloro-1-methylpseudouridine (03601015117):
Scheme 31
HN NH
HNANH
TBDMS-CI
HO TBDMSO ______________________________________________ BSA CH3CN
CH3I
imidazole
HO OH TBDMSO OTBDMS
1 79
0 0
HN AN/ HN AN/
-78 degree C
oCI
LDA, Br2/CDI4
TBDMSO TBDMSO ________________________________________________ TBAF
TBDMSO OTBDMS TBDMSO OTBDMS 0
83 92
HN AN
HO oCI
HO OH
93
Compound 92: N,N-Diisopropylamine (1.4 mL, 10 mmol) was dissolved in 20 mL of
anhydrous THF.
The solution was cooled to -78 C under nitrogen atmosphere. n-Butyl lithium
(4 mL, 10 mmol; 2.5 M
in hexane) was added dropwise under stirring over 1 h. A solution of compound
83 (2.2 g, 3 mmol) in
5 mL of anhydrous THF was added to the LDA solution prepared above. The
resulting reaction
mixture was stirred at -78 C for an additional 2 h. Bromine (5 mL) was
dissolved in 10 mL of
anhydrous carbon tetrachloride, and dried with molecular sieves. This bromine
solution was added to
the LDA solution of compound 83 under stirring at -78 C until pale yellow
color became orange. The
reaction mixture was stirred at -78 C for 30 min. TLC (PE-EA = 3:1) indicated
trace amount of
starting material left. While still cold, the reaction mixture was poured into
the mixture of sodium
thiosulfate and sodium bicarbonate aqueous solution. It was extracted with
ethyl acetate, and the
organic phase was dried over anhydrous sodium sulfate and filtered. The
solution was concentrated
under reduced pressure. The residue was purified by flash chromatography on a
silica gel column to
give 1.6g of 92.
6-Chloro-1-methylpseudouridine (93): 1.6 g of 92 obtained above was treated
with 10 mL of 0.5 M
TBAF solution in THF, and it was stirred at room temperature for 24 h and
concentrated. The residue
was purified by flash chromatography on a silica gel column using methylene
chloride-methanol
- 268 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
providing 120 mg product with 96.3% HPLC purity. It was characterized by NMR
and MS spectral
analysis to be the N1-methyl-6-chloro pseudouridine 93.
Example 39. Synthesis of 1-Benzyl-pseudouridine (03601015032):
Scheme 32
0
HNANH HNANH
Im,TBDMSCI CH3CN
0 _________________________________________ TBDMSO 1) BSA, 2.5 eq
HO 0
DMF 0
Benzyl
bromide
HO OH TBDMSO OTBDMS
1 79
0
0
HNAN HNAN
TBDMSO 0 TBAF/THF HO
____________________________________________ Jor 0
TBDMSO OTBDMS HO OH
94 95
Compound 94: Bis-trimethylsilylacetamide (BSA, 10 mL) was added to a stirred
solution of compound
79 (2.0 g, 3.4 mmol) in 20 mL of dichloromethane. After stirring for four hour
at 40 C, benzyl bromide
(0.5 mL) was added to the solution, and the solution was then heated at reflux
temperature for 5
days. The reaction mixture was concentrated to dryness under reduced pressure.
The residue was
purified via silica gel chromatography using gradient eluent PE:EA = 20:1-8:1
to give 1.4 g product 94
as a light yellow foam in 60.8% yield.
1-Benzyl-pseudouridine (95): To a solution of compound 94(1.4 g, leg) in THF
was added TBAF in
THF (1 M, 10 mL), and the mixture was stirred at room temperature overnight.
The mixture was
concentrated under reduced pressure to dryness. The crude product was purified
by silica gel
chromatography using MeOH:DCM = 1:50-1:25 giving 0.309 g desired product 95 as
a white solid in
51.0% yield. Purity: 97.9% (HPLC); 1H NMR (DMSO-d6) 6 11.41(s, 1H), 7.91 (s,
1H), 7.27-7.36 (m,
5H), 4.94-4.95 (d, 1H, J = 3.6Hz), 4.86 (s, 2H), 4.77-4.80 (t, 1H, J = 4.2Hz),
4.71-4.72 (d, 1H, J =
4.2Hz), 4.46-4.47 (d, 1H, J= 3.3Hz), 3.93-3.96 (m, 1H), 3.83-3.87 (m, 1H),
3.68-3.70 (m, 1H), 3.59-
3.63 (m, 1H), 3.42-3.47 (m, 1H); Mass Spectrum: 335.1 (M + H)+, 358.1 (M +
Na).
- 269 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 40. Synthesis of 1-Methyl-3-(2-N-t-Boc-amino-3-t-butyloxycarbonyl)
propyl
psudouridine (03601015036-Boc):
Scheme 33
H
HNANH NANH
HO TBDMS-CITBDMS0 1) HMDS, 2.5 eq, CH2CH2
:_D
imidazole 2) CH31, 1.5 eq;
refluxed
3 days; ¨80%
HO OH TBDMSO OTBDMS
pseudouridine
79
1
>,Op
0
>,-(DN0
0
HN N
Me
E BocNH
OH
BocNH
TBDMS0:_D
DIAD
PPh3 TBDMS0:_D
THF
TBDMSO OTBDMS
83 NaBH4 TBDMSO OTBDMS
98
CICOOEt, NMM TBAF
yOH
BocNH
0
96 yN)Nhne
BocNH CD
HO
HO OH
99
Synthesis compound 97: A solution of Boc-Asp(OtBu)-OH (96) (5.0 g, 17.3 mmol)
in 50 ml dry THF
was cooled to -10 degree C. N-Methylmorpholine (1.75 g, 17.3 mmol) was added.
After 1 min,
CICO2Et (1.65 ml, 17.3 mmol) was added dropwise. The reaction mixture was
stirred for an additional
min at -5 degree C. The precipitated N-methylmorpholie hydrochloride was
filtered off, and the
filtrate was added to a solution of NaBH4 (1.47 g, 38.9 mmol) in 20 mL of
water at 5-10 degree C
10 within 10 min. The reaction mixture was stirred at room temperature for
3.5 h and then cooled to 5
degree C. 3M hydrochloric acid was added to give a pH of 2, and the mixture
was extracted twice
with ethyl acetate. The combined organic phase was washed twice with water and
then dried with
anhydrous Na2504. The product is dried in vaccuo and purified via silica gel
chromatography using
EA: PE (1:2) as eluent to give 4.0 g product 97 as colorless oil in 85% yield.
Compound 98: Diisopropyl azodicarboxylate (1.6 g, 3 eq) was added to a stirred
solution of
compound 83(1.60 g, 1.0 eq), compound 97(0.83 g, 1.5 eq) and
triphenylphosphine (2.1 g, 3 eq) in
anhydrous THF (16 mL) at room temperature under N2. The reaction mixture was
stirred for 1 h, and
the solvent was removed under reduced pressure. The residue was purified by
silica gel
- 270 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
chromatography using PE: EA (10:1-8:1) providing 1.6 g desired product 98 as
pale yellow oil in
56.8% yield.
1-Methyl-3-(2-N-t-Boc-amino-3-t-butyloxycarbonyl) propyl psudouridine (99): To
a solution of
compound 98 (1.3 g, 1 eq) in THF was added TBAF in THF (1M, 2 mL), and the
mixture was stirred
at room temperature for 2 h. The mixture was concentrated to dryness under
reduced pressure. The
crude product was purified by silica gel chromatography using MeOH:DCM = 1:20-
1:5 to give 0.46 g
product 99 as white foam in 57.6% yield, with HPLC purity of 98%. 1H NMR (DMSO-
d6, 300MHz): 6
7.77 (5, 1H), 6.25-6.64 (d, 1H, J = 6.9 Hz), 4.93-4.95 (1H), 4.78-4.81 (m,
2H), 4.73-4.75 (d, 1H, J =
4.8 Hz), 4.51-4.52 (d, 1H, J= 2.4 Hz), 3.86-3.90 (m, 1H), 3.68-3.70 (m, 3H),
3.48 -3.50 (m, 3H), 3.34
(s, 3H), 2.33-2.37 (m, 2H), 1.26-1.37 (18H); ES MS, m/z 537.7 (M + Na).
Example 41. Synthesis of compound Pseudouridine 1-(2-ethanoic acid¨Fm)
(03601015034-
Fm):
Scheme 34
Br
Br" 1f' __________________________________________________
0
HO-Fm
0 0
/
HN NH HN NH Brif 0-Fm
'
Im,TBDMSCI 0 102
HO 0 TBDMS00o
DMF
1) BSA, 2.5 eq
CH3CN
HO OH TBDMSO OTBDMS
1 79
1110
0 INFO 0
HN
HN
o TBAF/THF 0
TBDMSO 0 HO 0
0
TBDMSO OTBDMS OH OH
100 101
Synthesis of Bromoacetic Acid Fm Ester (102): 9-Fluorenyl methanol (10 g, 1
eq) was dissolved in
100 mL of dichlormethane, and triethylamine (6.14 g, 1.19 eq) was added. The
reaction mixture was
cooled in ice bath, and a solution of bromoacetyl bromide (10.3 g, 1 eq) in 10
mL methylene chloride
was added under stirring over 1 h. The cloudy mixture was warmed to room
temperature, and stirred
overnight. The mixture was washed with water (100 mL x 3) and brine. The
combined organic phase
was dried and concentrated under reduced pressure. The crude product thus
obtained was purified
via silica gel chromatography (PE:EA = 300:1-50:1) giving 6.4 g product 102 as
a light yellow solid in
39.8% yield.
- 271 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Compound 100: Bis-trimethylsilylacetamide (BSA, 20 mL) was added to a stirred
solution of
compound 79 (2.5 g, 3.4 mmol) in dichloromethane (25 mL). After stirring for
four hours at 40 C, the
bromo-compound 102 (2.43 g, 1.8 eq) in dichloromethane (2 mL) was added, and
the solution was
then heated at reflux temperature for 5 days. The reaction mixture was
concentrated under reduced
pressure to dryness. The residue was purified via silica gel chromatography
using PE: EA=10:1-5:1
giving 0.8 g desired compound 100 as white foam. (1.7 g of compound 79 was
recovered).
Pseudouridine 1-(2-ethanoic acid ¨Fm) (101): To a solution of compound 100
(0.8 g, 1 eq) in THF (20
mL) was added 1M HCI (1 mL), and the mixture was stirred at room temperature
overnight. The
mixture was concentrated under reduced pressure to dryness. The residue was
purified by silica gel
chromatography using MeOH:DCM = 1:30-1:20 giving 0.30 g final product 101 as
white solid in 64.2
% yield.
Example 42. Synthesis of 5-Ethyl-cytidine (03601014039):
Scheme 35
0 HN)CCH3
Bz0 1,2,4-triazole
OAc + CH Bz0 0 N POCI3, 0 C
OBz OBz 0 N Et3N, MeCN
103 104 OBz OBz
105
Nr) NH2 NH2
N L.n3 N CH3
u
N
I
N NH3 H20 Bz0 0 N H3/Me0H HO 0
N
BzOAN
1,4-Dioxane
OBz OBz OH OH
OBz OBz 108
107
106 [5-Ethyl-
cytidine]
Compound 105: To a solution of compound 104 (2.8 g, 20 mmol) in dry
acetonitrile (30 mL) was
added BSA (21 g, 100 mmol, 5 eq). The reaction mixture was stirred at 60 C
for 4 h and cooled to
room temperature. To this reaction mixture were added compound 103 (10.1 g, 20
mmol), TMSOTf
(10.8 mL, 60 mmol, 3eq), and the resulted reaction mixture was stirred at 60
C for 4 h. Upon
completion of the reaction as monitored by TLC, the reaction mixture was
treated with methylene
chloride and saturated sodium bicarbonate. The organic phase was separated,
and the aqueous
phase was extracted with dichloromethane. The combined organic phase was dried
over anhydrous
Na2SO4. The drying agent was filtered off, and the filtrate was concentrated
under reduced pressure.
The crude product was purified by flash chromatography on a silica gel column
giving 11 g desired
compound 105 in 95% yield.
- 272 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Compound 106: To a solution of 1,2,4-1H-triazole (19.36 g, 285 mmol),
phosphorus oxychloride (5.8
mL 63 mmol) in dry methylene chloride (300 mL) was added slowly triethylamine
(37.5 mL, 270
mmol) at 0 C. After the reaction mixture was warmed to room temperature,
compound 105 (16.7 g,
30 mmol) was added. The reaction mixture was added and stirred at temperature
for 2 h. Upon
completion of the reaction as monitored by TLC, the reaction mixture was
treated with methylene
chloride and saturated sodium bicarbonate. The organic phase was separated,
and the aqueous
phase was extracted with methylene chloride. The combined organic phase was
dried over
anhydrous Na2SO4. The drying agent was filtered off, and the filtrate was
concentrated under reduced
pressure giving crude product compound 106 which was carried to the next step
without further
purification.
Compound 107: To a stirred solution of compound 106 (crude obtained above) in
dioxane (135 mL)
was added concentrated ammonia solution (19.4 mL). The reaction mixture was
stirred at room
temperature for 5 h. Upon completion of the reaction as monitored by TLC, the
reaction mixture was
concentrated under reduced pressure to give crude compound 107 which was
carried to the next step
without further purification.
5-Ethyl-cytidine (108): A solution of compound 107 (crude obtained above) in
saturated ammonia
methanol solution (100 mL) was stirred at room temperature ins sealed
container for 24 h. Upon
completion of the reaction as monitored by TLC, the reaction mixture was
concentrated under
reduced pressure to dryness. The crude product was purified by flash
chromatography on a silica gel
column resulting in the desired final product 108 which was was characterized
by NMR, MS and UV
spectral analyses. 1H NMR (DMSO-d6) 6 7.8 (s,1H), 7.3 (brs, 2H), 5.75 (s, 1H),
5.31 (s, 1H), 5.16 (s,
1H), 5.01 (s, 1H), 3.97 (s, 2H), 3.83 (s, 1H), 3.68 (d, 2H, J = 12.0 Hz), 3.55
(d, 2H, J = 12.4 Hz), 2.25
(q, 2H, J = 7.2 Hz), 1.05 (t, 3H, J = 7.2 Hz). Mass Spectrum: m/z 272.0 (M +
H) .
- 273 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 43. Synthesis of 5-Methoxy-cytidine (03601014030):
Scheme 36
OCH3
HN 1,2,4-triazole
Bz0
0 )-OCH3
_______________________________________________________ Bz0( )N POCI3, 0 C
0Ac + HN
Et3N, MeCN
OBz OBz
103 109 OBz OBz
110
NH2 NH2
OCH3 N
OCH3
N
H3C0
NH3 H20 Bz0(113N NH3/Me0H
HO 00 N
BzON 0 1,4-Dioxane
OBz OBz OH OH
OBz OBz 113
111
112 [5-Mehoxy-
cytidine]
Compound 110: To a solution of compound 109 (1.42 g, 10 mmol) in dry
acetonitrile (30 mL) was
added BSA (10.5 g, 50 mmol). The reaction mixture was stirred at 60 C for 4 h
and cooled to room
temperature. To the reaction mixture were added compound 103 (5.04 g, 10 mmol)
and TMSOTf (2.7
mL, 15 mmol). The resulted reaction mixture was stirred at 60 C for 4 h. Upon
completion of the
reaction as monitored by TLC, the reaction mixture was treated with methylene
chloride and
saturated sodium bicarbonate. The organic phase was separated, and the aqueous
phase was
extracted with methylene chloride. The combined organic phase was dried over
anhydrous Na2SO4.
The drying agent was filtered off, and the filtrate was concentrated under
reduced pressure. The
crude product was purified by flash chromatography on a silica gel column
giving 3.8 g desired
compound 110 in 65% yield.
Compound 111: To a solution of 1,2,4-1H-triazole (8.73 g, 126 mmol) and
phosphorus oxychloride
(2.6 mL 27.9 mmol) in dry methylene chloride (300 mL) was added slowly
triethylamine (16.6 mL,
119.8 mmol) at 0 C . After the reaction mixture was warmed to room
temperature, compound 110
(7.8 g, 13.3 mmol) was added. The reaction mixture was stirred at temperature
for 2 h. Upon
completion of the reaction as monitored by TLC, the reaction mixture was
treated with methylene
chloride and saturated sodium bicarbonate. The organic phase was separated,
and the aqueous
phase was extracted with methylene chloride. The combined organic phase was
dried over
anhydrous Na2SO4. The drying agent was filtered off, and the filtrate was
concentrated under reduced
pressure giving crude product compound 111 which was carried to the next step
without further
purification.
Compound 112: To a stirred solution of compound 111 (crude obtained above) in
dioxane (60 mL)
was added concentrated ammonia solution (8.6 mL). The reaction mixture was
stirred at room
temperature for 5 h. Upon completion of the reaction as monitored by TLC, the
reaction mixture was
- 274 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
concentrated under reduced pressure giving crude compound 112 which was
carried to the next step
without further purification.
5-Methoxy-cytidine (113): A solution of compound 112 (crude obtained above) in
saturated ammonia
methanol solution (80 mL) was stirred at room temperature in a sealed
container for 24 h. Upon
completion of the reaction as monitored by TLC, the reaction mixture was
concentrated under
reduced pressure to dryness. The crude product was purified by flash
chromatography on a silica gel
column resulting in the desired final product 113 which was characterized by
LC-MS, UV and HNMR.
1H NMR (DMSO-d6) 6 7.73 (s,1H), 7.50 (s, 1H), 7.03 (s, 1H), 5.76 (d, 1H, J =
3.6 Hz), 5.31 (s, 1H),
5.26 (d, 1H, J = 4.0 Hz), 4.96 (d, 1H, J = 4.8 Hz), 4.01 (d, 1H, J = 4.4 Hz),
3.95 (s, 1H), 3.83 (d, 1H, J
= 2.8 Hz), 3.78 (d, 1H, J= 12.0 Hz), 3.62 (s, 3H), 3.58 (d, 1H, J= 12.4 Hz);
Mass Spectrum: m/z
274.0 (M + H) .
Example 44. Synthesis of 2-Thio-5-amino(TFA)-methyl-Uridine (00901013015-TFA):
Scheme 37
o o
o
Ac0 HN HN
, Ao-
I j ________
HN)
0 OAc 1) HMDS s')'--N--. Na0Me S N
.-
1 ___________________________________ .-
S N MO OM 2 SnCI4 r..- Ac0:_D
H or K2CO3 Ts0H, acetone
DMF
114 115
Ac0 OAc OH OH
116 117
0 0 0
HN)c HNOH
HNCI
j
TMSCI I
j
S N (HCNO) n S N 5 eq S N
___________________________________________________ .- HO
HO HO NH3 H20
_?0
heat ___________________________________________________________________ .-
NaOH
heat
00 070
00
118 119 /\ 120
0 0 0
---11---..7'NH2 NH
HN 1 HN-"H HI\I-jir
HO SN% 1
3 acid
0.'''CF3
(CF3C0)20 HO SN 0 CF HO S'N1-.'
_______________________________ ..-
1-r
07<.,0 OXD OH OH
123
121 122
Compound 116: A mixture of 2-thiouracil 114 (6.0 g, 46.8 mmol), trimethyl
chlorosilane (5.4 mL),
hexamethyldisilazane (240 mL) and catalytic amount of ammonium sulfate were
refluxed for 18 h.
Upon the reaction mixture became clear, it was concentrated under reduced
pressure to dryness at
the temperature not greater than 45 C. To the resulted silylated thiouracil
was dissolved in 1,2-
dichloroethane (60 mL), and 1,2,3,5-tetra-0-acetyl-D-ribofuranose (16.5 g,
51.9 mmol) was added. It
was stirred until homogeneous, stannic chloride (7.2 mL, 62.4 mmol) was added
and stirred or 1 h.
Upon completion of the reaction as monitored by TLC, the reaction mixture was
poured into 150 mL
of saturated sodium bicarbonate and stirred for 1 h. The mixture was filtered
through a pad of Celite,
- 275 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
and washed with methylene chloride. The organic phase was separated, and the
aqueous was
extracted with dichloromethane. The combined organic phase was dried over
anhydrous Na2SO4.
The drying agent was filtered off, and the filtrate was concentrated under
reduced pressure. The
crude product was purified by flash chromatography on a silica gel column
using ethyl acetate -
petroleum ether (1:2 to 1:1) resulting in compound 116 (15.0 g, 38.8 mmol) in
82.9% yield.
Compound 117: To a stirred solution of compound 116 (15.0 g, 38.8 mmol) in
absolute methanol (150
mL) was added lithium hydroxide (3.7 g, 155.2 mmol, 4 eq), and the reaction
mixture was stirreed at
room temperature for 30 min. Upon completion of the reaction as monitored by
TLC, hydrochloric acid
(3 N) was added to adjust to neutral. The mixture was concentrated under
reduced pressure resulting
in the white precipitate which was filtered giving 5 g of desired product. The
filtrate was concentrated
under reduced pressure to give crude product. The crude product was purified
by flash
chromatography on a silica gel column using methylene chloride-methanol (10:1
to 5:1) resulted
compound 117 (1.2 g). 6.2 g (23.8 mmol) in 61.3% yield.
Compound 118: To a stirred solution of compound 117 (6.0 g, 23.1 mmol) in
acetone (60 mL) was
added p-toluenesulfonic acid (0.8 g, 4.7 mmol) and 2,2-Dimethyoxypropane (5.0
g 48.1 mmol). The
resulted reaction mixture was stirred at room temperature for 2 h, and solid
material disappeared.
Upon completion of the reaction as monitored by TLC, sodium bicarbonate (1.5
g) was added, and it
was stirred for 1 h. The solid was filtered off and washed with
dichloromethane. The filtrate was
concentrated under reduced pressure. The crude product was purified by flash
chromatography on a
silica gel column using methylene chloride-methanol (20:1 to 10:1) as eluent
resulting in (6.4 g, 21.3
mmol) compound 118 in 92.2% yield.
Compound 119: To a stirred solution of compound 118 (6.0 g, 20 mmol) in
aqueous potassium
hydroxide (0.5 M, 100 mL) was added paraformaldehyde (3.0 g, 100 mmol). The
resulted reaction
mixture was stirred at 50 C overnight. Upon completion of the reaction as
monitored by TLC,
hydrochloric acid (3 M) was added to adjust to neutral. The mixture was
concentrated under reduced
pressure to dryness. The crude product was purified by flash chromatography on
a silica gel column
using methylene chloride-methanol (20:1 to 10:1) resulting in (4.2 g, 12.7
mmol) compound 119 in
63.6 % yield.
Compound 120: To a stirred solution of compound 119 (7.5 g, 22.7 mmol) in
dioxane (50 mL) was
added TMSCI (14.5 mL, 113 mmol, 5 eq). The reaction mixture was stirred at 50
C under N2
atmosphere overnight. Upon almost completion of the reaction as monitored by
TLC, the reaction
mixture was concentrated at the temperature not over 30 C under reduced
pressure. The crude
product was dissolved in anhydrous acetone, and concentrated under reduced
pressure to dryness.
Thus resulted crude product compound 120 was used in next step without further
purification.
Compound 121: To a stirred solution of compound 120 (crude obtained above) in
dioxane (50 mL)
was added ammonium hydroxide. The reaction mixture was stirred at room
temperature overnight.
- 276 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Upon completion of the reaction as monitored by TLC, the reaction mixture was
concentrated under
reduced pressure to dryness. The crude product was purified by flash
chromatography on a silica gel
column using ethyl acetate - petroleum ether (1:3 to 1:1) as eluent resulting
in compound 121 (3.1 g)
which was used in next step directly.
Compound 122: A solution of compound 121 (3.1 g, 7 mmol) in dry pyridine (50
mL) was cooled to 0
C, and trifluoroacetic anhydride (18 g, 8 mmol) was added under N2 atmosphere.
The reaction
mixture was stirred at room temperature for 1 h. Upon completion of the
reaction as monitored by
TLC, the reaction mixture was diluted with methylene chloride (100 mL) and
aqueous sodium
bicarbonate (100 mL, 5 %). The organic phase was separated, and the aqueous
phase was extracted
with dichloromethane. The combined organic phase was dried over anhydrous
Na2SO4. The drying
agent was filtered off, and the filtrate was concentrated under reduced
pressure to dryness. The
crude product was purified by flash chromatography on a silica gel column
using ethyl acetate -
petroleum ether (1:5 to 1:3) as eluent resulting in compound 122 which was
used directly in next step.
2-Thio-5-amino(TFA)-methyl-Uridine (123): 10 mL of hydrochloric acid (1 M) was
added to a flask
containing compound 122 (1.0 g). The mixture was stirred at room temperature
for 30 min. The
reaction mixture was neutralized with Na2CO3. The solid was filtered off, and
the filtrate was
concentrated under reduced pressure to dryness. The crude product was purified
by flash
chromatography on a silica gel column giving 290 mg desired final compound
123. Compound 123
was characterized by NMR, MS and UV with 99.0% HPLC purity: 1H NMR (DM50-d6) 6
12.73 (s,1H),
9.56 (s, 1H), 8.17 (s, 1H), 6.57 (s, 1H), 5.42 (d, 1H, J = 4.8 Hz), 5.17 (s,
1H), 5.12 (d, 1H, J = 4.4 Hz),
4.02-3.97 (m, 4H), 3.92 (s, 1H), 3.71 (d, 1H, J= 12.0 Hz), 3.60 (d, 1H, J= 6.6
Hz); Mass Spectrum:
m/z 385.7 (M + H)+; 407.7 (M+Na) .
- 277 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Example 45. Synthesis of 5-Formy1-2'-0-methylcytidine (03601074036):
Scheme 38
NH2 NH2
H3CN
HO TBSO N 0 1,2,4-triazole
TBSCI POCI3, 0 C
1¨r imidazole
1¨r Et3N, MeCN
OH OCH3 0 OCH3
TBS-
124 125
r¨N
NH2
H3C
N
TBSO
N 0 1,4-Dioxane TBSO NO 2, 6-
_____________________________________ * lutidine
NH4OH
1¨r CuSO4
TBS0 OCH3 TBS 0 OCH3 Na2S208
-
-
126 127
NH2 NH2
OHC OHC
N N
TBSO
TBAF
N 0 HO N 0
1¨r THF
1¨r
TBS0 OCH3 OH OCH3
-
128 129
Compound 125: To a solution of compound 124 in dry N,N-dimethylformamide were
added tert-
Butyldimethylsilyl chloride (3 eq) and imidazole (4 eq). The reaction mixture
was stirred at room
temperature overnight and then quenched with water. The mixture was extracted
with ethyl acetate,
and the combined organic phase was washed with brine, and dried over anhydrous
Na2SO4. The
drying agent was filtered off, and the filtrate was concentrated to dryness
under reduced pressure.
The crude product thus obtained was purified by flash chromatography on a
silica gel column giving
compound 125.
Compound 126: To a solution of 1,2,4-1H-triazole (4.58 g, 66.3 mmol) in dry
methylene chloride (500
mL) was added slowly phosphorus oxychloride (1.34 mL, 14.4 mmol) at room
temperature. The
mixture was cooled to 0 C, and triethylamine (8.7 mL) was added followed by
the addition of
compound 125 (3.5 g, 7 mmol) in dichloromethane. The reaction mixture was
allowed to warm to
room temperature, and stirred for 30 min. Upon completion of the reaction as
monitored by TLC, the
reaction mixture was treated with a mixture of triethylamine and water,
followed by addition of
saturated sodium bicarbonate. The organic phase was separated, and dried over
anhydrous Na2SO4.
The drying agent was filtered off, and the filtrate was concentrated under
reduced pressure giving
crude product compound 126 which was carried to the next step without further
purification.
- 278 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Compound 127: To a stirred solution of compound 126 (crude obtained above) in
dioxane (25 mL)
was added concentrated ammonium solution (4 mL). The reaction mixture was
stirred at room
temperature for 1 h. Upon completion of the reaction as monitored by TLC, the
reaction mixture was
concentrated under reduced pressure giving crude compound 127. The crude
product was purified by
flash chromatography on a silica gel column using methanol- dichloromethane
(1:10) as eluent
providing desired product 127.
Compound 128: To a stirred solution of compound 127 (5 g) in acetonitrile (70
mL) were added 2,6-
lutidine (3.7 g), and an aqueous solution of sodium persulfate (4.76 g, 20 mL)
and copper sulfate
(0.638 g, aq. solution). The reaction mixture was stirred at 60 C for 2 h.
The mixture was extracted
with dichloromethane. The organic phase was washed with brine and dried over
anhydrous Na2SO4.
The drying agent was filtered off, and the filtrate was concentrated to
dryness under reduced
pressure. The crude product thus obtained was purified by flash chromatography
on a silica gel
column giving desired compound 128.
5-Formy1-2'-0-methylcytidine (129): To a stirred solution of compound 128 (1
g, 2 mmol) in dry
tetrahydrofuran (15 mL) were added a solution of tetrabutylammonium fluoride
in tetrahydrofuran (1
M), followed by the addition of acetic acid (0.3 eq). The reaction mixture was
stirred at room
temperature. Upon completion of the reaction as monitored by TLC, the reaction
mixture was
concentrated under reduced pressure to dryness. The crude product was purified
by flash
chromatography on a silica gel column giving desired compound 129 with 99%
HPLC purity.
Compound 129 was characterized by NMR, MS and UV. 1H NMR (DM50-d6) 6 9.39
(s,1H), 9.04 (s,
1H), 8.16 (s, 1H), 7.84 (s, 1H), 5.83 (s, 1H), 5.32 (s, 1H), 5.08 (d, 1H, J =
6.4 Hz), 4.10 (d, 1H, J = 4.8
Hz), 3.89 (d, 1H, J = 6.8 Hz), 3.81 (s, 1H), 3.74 (s, 1H), 3.64 (d, 1H, J =
5.0 Hz), 3.32 (s, 3H); Mass
Spectrum: m/z 286 (M + H)+; 571 (2M+H) .
- 279 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Example 46. Synthesis of 2'-0-Methyl-2-thiouridine (00901073008):
Scheme 39
NH
NH
HO NO MsCI, -78 degree Ms-0 N 0 NaHCO3,
Et0H
0
Py, 0 degree reflux 36 h
OH OCH3 OH OCH3
130 131
AN
NH
N
HO
H2S saturated at -50 degree S
HO
0 0
py, bump, sealed
heat 50-60 degree
O
OH OCH3 H OCH3
1
133 34
[2-Thio-2-0-methyluridine]
Compound 131: A solution of compound 130 (5.16 g, 20 mmol) in dry pyridine
(100 mL) was cooled
to -78 C, and MsCI (1.86 mL, 2.76 g, 24 mmol, 1.2 eq) was added dropwise. The
reaction mixture
was allowed to warm to room temperature, and continued to stir for 1 h. Upon
completion of the
reaction as monitored by TLC, the reaction mixture was quenched with methanol
(1 mL), and
concentrated under reduced pressure. The crude product was purified by flash
chromatography on a
silica gel column using dichloromethane - methanol (50:1 to 20:1) resulting in
compound 131 (3.4 g,
10 mmol) in 50% yield.
Compound 133: A mixture of compound 131 (3.36 g, 10 mmol) and sodium
bicarbonate (2.1 g, 25
mmol) in ethanol (250 mL) was refluxed under N2 atmosphere for 36 h. The
reaction mixture was
cooled to room temperature, and solid sodium bicarbonate was filtered off. The
filtrate was
concentrated under reduced pressure, and the crude product was purified by
flash chromatography
on a silica gel column using dichloromethane -methanol (50:1 to 20:1)
resulting in 1.7 g of compound
133 in 59% yield. Some starting material was recovered. This product was
verified by MS spectrum
with good HPLC purity.
2'-0-Methyl-2-thiouridine (134): A solution of compound 133 (1.7 g, 5.94 mmol)
in 500 mL of
anhydrous pyridine in a high-pressure bump vessel was cooled to -50 C. The in
house prepared and
dried hydrogen sulfide gas was bubbled in the solution to make it saturated at
low temperature. The
high-pressure bump was sealed, and heated in an oil bath to 50 C for 4 h, and
then increased to 70
C for 24 h. The reaction vessel was cooled to room temperature, and allowed to
open to the air
slowly. The reaction mixture was concentrated under reduced pressure, and the
residue was purified
by flash chromatography on a silica gel column providing desired final product
134 with 98.79%
HPLC purity (some starting material was recovered). It was characterized by
NMR, MS and UV. 1H
NMR (DM50-d6) 6 12.66 (s,1H), 8.20 (d, 1H, J= 8.0 Hz), 6.60 (d, 1H, J= 3.2
Hz), 6.00 (d, 1H, J=
- 280 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
8.4 Hz), 5.28 (d, 1H, J= 4.8 Hz), 5.17 (d, 1H, J= 6.0 Hz), 4.10 (t, 1H, J= 5.2
Hz), 3.90 (d, 1H, J= 3.2
Hz), 3.80 (d, 1H, J= 4.4 Hz), 3.75 (d, 1H, J= 4.0 Hz), 3.62 (d, 1H, J= 4.0
Hz), 3.45 (s, 3H); Mass
Spectrum: m/z 275 (M + H)+; 297 (M+Na) .
Example 47. Synthesis of 2-Selenouridine (03601013046):
Scheme 40
HN ,
HN
Ac0 ,
HN (: c 1) HMDS _?),OA sN Me0H SN
________________________________________ AcO HO
Ac0 OAc ,
S N 2 SnCI4 LiOH
1¨r
114 115
Ac0 OAc OH OH
116 117
0 0
HN N
1)NaBH4,Se,ethanol
TBDMSCI
SN DBU,Mel
30min 0
______________ -TBDMSO TBDMSO S N
imidozle 0 DMF 0 2) ethanol
DMF 3 d rt.
TBDMSO OTBDMS TBDMSO OTBDMS
135 136
0 0
HN HN
Se TBAF
TBDMSO HO SeN
THF
1¨r 1¨r
TBDMSO OTBDMS OH OH
137 138
Compound 135: A solution of compound 117 (12 g, 46.1 mmol), t-
butyldimethylsilyl chloride (70 g,
461.0 mmol, 10 eq), and imidazole (36.55 g, 553.2 mmol, 12 eq) in 150 mL of
anhydrous DMF was
stirred at 60 C for 12 h. Upon completion of the reaction as monitored by
TLC, the reaction mixture
was quenched with water and extracted with dichloromethane. The organic phase
was dried over
anhydrous sodium sulfate, and concentrated under reduced pressure. The residue
was purified by
flash chromatography on a silica gel column providing 20 g compound 135 in 72%
yield.
Compound 136: To a solution of compound 135 (5 g, 8.3 mmol) in 50 mL of
anhydrous DMF was
added iodomethane (11.8 g, 83 mmol, 10 eq), followed by addition of DBU (1.9
g, 12.45 mmol, 1.5
eq). The reaction mixture was stirred at room temperature for 12 h, and
quenched with water. The
mixture was extracted with dichloromethane. The organic phase was dried over
anhydrous sodium
sulfate, and concentrated under reduced pressure. The residue was purified by
flash chromatography
on a silica gel column providing 2.0 g compound 136 in 39% yield.
- 281 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Compound 137: A suspension of selenium (1.28 g, 16.2 mmol, 5 eq) and sodium
borohydride (0.74 g,
19.44 mmol, 6 eq) in anhydrous ethanol was stirred at 0 C under nitrogen flow
for 30 minutes till
clear colorless solution. A solution of compound 136 (2.0 g, 3.24 mmol) in 10
mL of ethanol was
added to the selenium hydride system with syringe. The reaction mixture was
stirred at room
temperature for 3 days and monitored by TLC. It was quenched with water and
extracted with
methylene chloride. The organic phase was dried over anhydrous sodium sulfate
and concentrated
under reduced pressure. The residue was purified by flash chromatography on a
silica gel column
providing 1.8 g product 137 in 85% yield.
2-Selenouridine (138): To a solution of compound 137 (1.8 g, 2.7 mmol) in 10
mL of THF was added
17 mL of TBAF solution in THF (1 mol/L). It was stirred at room temperature
for 2 hours. The reaction
mixture was quenched with water and concentrated under reduced pressure to
dryness. The residue
was purified several times by flash chromatography on silica gel columns
providing 260 mg of
compound 138 with 96% HPLC purity. It was characterized by NMR, MS and UV
spectral analyses.
1H NMR (DMSO) 6 13.9 (s,1H), 8.21 (d, J = 8.0Hz, 1H), 6.70 (d, J = 4.0 Hz,
1H), 6.13 (d, J = 8.4 Hz,
1H), 5.42 (d, J = 5.2Hz, 1H), 5.26 (t, J = 4.4Hz, 1H), 5.11 (d, J= 5.6Hz, 1H),
4.11-4.06 (m, 1H), 4.01-
3.95 (m, 1H), 3.94-3.90 (m, 1H), 3.75-3.67 (m, 1H), 3.64-3.56 (m, 1H). Mass
Spectrum: m/z 308.8 (M
+ H) . 330.7 (M + Na).
- 282 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Example 48. Synthesis of 5-N-methyl-N-TFA-aminomethy1-2-thiouridine
(00901013015-N-Me,N-
TFA):
Scheme 41
0
0 )"
Bz0
HN
OAc 1) HMDS 1
SNz deprotect
HN I
_________________________________________ , __ Bz0 _______________ ,
SNz +
Bz0 OBz 2 SnCI4
0 LiOH
H
114 103
Bz0 OBz
139
0 0 0
N /0¨ )" HN )-
1 OH
HN 1 HN 1
HO
SN ____ HO A
0- SN z (HCHO)n HO
SN7
, _____________________________________________________ ' 0
Ts0H, acetone NaOH
DMF heat
OH OH 070 070
117
1
140 41
0 0
k z Me
HN I CI HN- 'NH
TMSCI
SN (CF3C0)20
eq SN NH Me
HO
_________________ - HO 2 , ______________________________
,..-
heat (:::_i 0
water-dioxane
C50 070
142 143
0 0
)cv CH3 )c7 CH3
HN 1 N HN N
HO
SN 0CF3 acid
HO SNj 0CF3
,-
00 OH OH
/
K. 144
145
5 Synthesis of Compound 139. To a solution of compound 114 (24.0 g, 187.2
mmol) in
hexamethyldisilazane (960 mL) were added trimethyl chlorosilane (21.60 mL,
169.70 mmol) and the
catalytic amount of ammonium sulfate (1.0 g, 75 mmol). The clear reaction
mixture was stirred at 126
C for 18 h. The reaction mixture became clear, and concentrated under reduced
pressure to dryness
at not more than 45 C. A solution of 1,2,3,5-tetra-0-acetyl-D-ribofuranose
(66 g, 207.60 mmol) in dry
1,2-dichloroethane (240 mL) was added to the reaction mixture, followed by
addition of tin
tetrachloride (28.80 mL, 249.6 mmol). The reaction mixture was stirred at room
temperature for 1 h.
Upon completion of the reaction as monitored by TLC, the reaction mixture was
poured into saturated
sodium bicarbonate (1000 mL) and stirred for 1 h. The solid was filtered off
through a pad of Celite,
- 283 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
and washed with methylene chloride. The organic phase separated, and the
aqueous phase was
extracted with dichloromethane. The combined organic phase was dried over
anhydrous Na2SO4.
The drying agent was filtered off, and the filtrate was concentrated under
reduced pressure. The
crude product was purified by flash chromatography on a silica gel column
using ethyl acetate -
petroleum ether (1:2 to 1:1) resulting in compound 139 (65.0 g, 168.2 mmol) in
89% yield.
Synthesis of Compound 117. To a stirred solution of compound 139 (70 g, 181.17
mmol) in methanol
(700 mL) was added lithium hydroxide (15 g, 625 mmol). It was stirred at room
temperature for 1 min.
Upon completion of the reaction as monitored by TLC, the reaction mixture was
treated with
hydrochloric acid (3 N) to adjust to neutral. The reaction mixture was
concentrated under reduced
pressure resulting in white solid precipitation. The precipitate was filtered
giving 31.2 g compound 117
as white solid in 66.2 % yield.
Synthesis of Compound 140. To a stirred solution of compound 117 (31.20 g,
119.88 mmol) in dry
acetone (1000 mL) were added p-toluenesulfonic acid (3.06 g, 17.79 mmol) and
2,2-
dimethoxypropane. The resulted reaction mixture was stirred at room
temperature for 2 h till solid
completely disappeared. Upon completion of the reaction as monitored by TLC,
the reaction mixture
was adjusted to neutral with by addition of saturated sodium bicarbonate (150
mL). The solid was
filtered off, and washed with dichloromethane. The filtrate was concentrated
under reduced pressure,
and the crude product was purified by flash chromatography on a silica gel
column using
dichloromethane -methanol (20:1 to 10:1) to give final product compound 140
(33.97 g, 113.10 mmol)
in 94.36 % yield.
Synthesis of Compound 141. To a stirred mixture of compound 140 (26.0 g, 86.57
mmol) and
aqueous sodium hydroxide (0.5 N, 200 mL) was added paraformaldehyde (20.0 g,
666.66 mmol). The
reaction mixture was stirred at 50 C overnight. Upon completion of the
reaction as monitored by
TLC, the reaction mixture was adjusted to neutral with hydrochloric acid (3
N). The reaction mixture
was concentrated under reduced pressure to dryness. The crude product was
purified by flash
chromatography on a silica gel column using methylene chloride- methanol (20:1
to 10:1) resulting in
compound 141 (25 g, 75.76 mmol) in 87.51 % yield.
Synthesis of Compound 142. Compound 141 (16 g, 48.43 mmol) was dissolved in
anhydrous dioxane
(500 mL), and chlorotrimethylsilane (65 mL, 507 mmol) was added to the stirred
solution. The
reaction mixture was stirred overnight at 50 C under N2 atmosphere. The
reaction mixture was
concentrated under reduced pressure at not less than 30 C giving crude
product compound 142
which was carried to the next step without further purification.
Synthesis of Compound 143. To a stirred solution of compound 142 (crude
obtained above) in
dioxane (200 mL) was added methylamine MeNH2 (200 mL, 40% aq. Solution, 2.32
mol, 48 eq). The
reaction mixture was stirred at room temperature for 10 min. Upon completion
of the reaction as
monitored by TLC, the reaction mixture was concentrated under reduced pressure
to dryness. The
- 284 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
crude product was purified by flash chromatography on a silica gel column
using methylene chloride-
methanol (30:1 to 20:1) resulting in compound 143 (6.7 g 19.51 mmol) in 40.28%
yield.
Synthesis of Compound 144. To a stirred solution of compound 143 (6.45 g,
18.78 mmol) in dry
pyridine (100 mL) was added trifluoroacetic anhydride (7.94 mL, 56.32 mmol, 3
eq). The reaction
mixture was stirred at room temperature for 10 h. Upon completion of the
reaction as monitored by
TLC, the reaction mixture was concentrated under reduced pressure to dryness.
The crude product
was purified by flash chromatography on a silica gel column using ethyl
acetate - petroleum ether (1:5
to 1:2) resulting in compound 144 (7.5 g, 17.06 mmol) in 90.84% yield.
Synthesis of Compound 145. To a stirred solution of compound 144(6 g, 13.65
mmol) in methanol
(60 mL) was added hydrochloric acid (1 N, 35 mL). It was stirred at room
temperature for 10 h, and
then stirred at 80 C for 0.5 h. Upon completion of the reaction as monitored
by TLC, the reaction
mixture was cooled to room temperature and treated with methylene chloride (10
mL). The reaction
mixture was concentrated under reduced pressure to dryness. The crude product
was purified by
flash chromatography on a silica gel column using methylene chloride-methanol
(30:1 to 20:1)
resulting in 2.5 g of final product 145 in 45.86% yield with 99.29% HPLC
purity. Compound 145 was
characterized by NMR, MS and UV spectral analyses. 1H NMR (DM50-d6) 6 s,1H),
8.2 (d, J = 6.9Hz,
1H), 6.5 (t, J= 2.1Hz, 1H), 5.4 (d, J= 3.9Hz, 1H), 5.20-5.07 (m, 2H), 4.37-
4.15 (m, 2H), 4.08-4.05
(dd, 1H), 3.99-3.91 (m, 2H), 3.75-3.57 (m, 2H), 3.1 (d, J= 1.2Hz, 2H), 2.9 (s,
1H). Mass Spectrum
m/z 400 (M + H) . 422 (M + Na). UV, Amax = 278 nm.
Example 49. Synthesis of 5-(Ethoxycarbonyl)(hydroxy)methy1-2',3',5'-tris-0-
(tert-
butyldimethylsilyOuridine (03601013047):
Scheme 42
0 0
0
)Br
HN)C, HN HN
HO ON, HO 0 N __________________________________________ TBDMSO 0 N
OH OH OH OH
TBDMSO OTBDMS
146 147 148
OH 0 OHO
NH 1M HCI NH
0 I
Me0H 0 I
0 N 0 _______________________
NO
BuLi, THF ethyl glyoxylate TBDMSO HO
-78 C -78 C
TBDMSO OTBDMS OH OH
149 150
- 285 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Synthesis of Compound 147: To a solution of uridine 146 (20.0 g, 82mmol) and
NBS (21.7 g, 0.12
mol) in anhydrous dimethylfomamide was added AIBN (0.1 eq) in anhydrous
dimethylfomamide, then
the solution was stirred at 80 C for 4 h. Saturated sodium thiosulfate
solution (20 mL) was added.
After evaporation of the solvent, the residue was precipitation with methanol
to give 22.0 g compound
147 as light yellow solid.
Synthesis of Compound 148: To the solution of compound 147 (22.0 g, 66 mmol),
imidazole (23.0 g,
0.33 mol) in anhydrous dimethylfomamide (100 mL) was added TBDMSCI (50.0 g,
0.32 mol) in
anhydrous dimethylfomamide (50.0 mL), then the solution was stirred at it
overnight. Saturated
sodium bicarbonate solution (30 mL) was added. The aqueous phase was extracted
with ethyl
acetate (2 x 300 mL), and the combined organic phase was washed with brine,
and dried over
sodium sulfate. After evaporation of the solvent, the residue was purified by
silica gel chromatography
giving 40.0 g compound 148 as light yellow syrup.
Synthesis of Compound 149: To a solution of compound 148 (10.0 g, 15.0 mmol)
in anhydrous THF
(100 mL) at -78 C was added n-BuLi (2.5 M in hexane, 24 mL). The solution was
stirred for 1 h, and
freshly distilled ethyl glyoxylate (32 mmol) was added. The mixture was
stirred for 1 h at -78 C,
warmed to room temperature, and stirred overnight. Saturated ammonium chloride
(50 mL) was
added. The aqueous phase was extracted with ethyl acetate (3 x 100 mL), and
the combined organic
phase was washed with brine, and dried over sodium sulfate. After evaporation
of the solvent, the
residue was purified by silica gel chromatography, eluting with 1-3% methanol
in dichloromethane,
giving 4.0 g compound 149 as light yellow syrup.
Synthesis of Compound 150: 5-(Ethoxycarbonyl)(hydroxy)methy1-2',3',5'-tris-0-
(tert-
butyldimethylsilyl)uridine compound 149 (4.0 g, 5.8 mmol) was treated added
HCI saturated solution
in methanol (0.5 M, 50 mL). The mixture was stirred at room temperature
overnight. After
concentrating the mixture to dryness under reduced pressure, the residue was
purified by silica gel
chromatography, eluting with 8-12% methanol in dichloromethane, giving
compound 150 as light
yellow foam. HPLC purity: 96%. 1H NMR (300MHz, DMSO-d6) 6 11.46 (s, 1H), 7.89-
7.93 (m, 1H),
5.80-5.86 (m, 2H), 5.39 (s, 1H), 5.06-5.12 (m, 2H), 4.83 (s, 1H), 3.55-3.95
(m, 8H); ESI mass
spectrum m/z: 332.8 [M + H], 254.8 EM + Na]. UV, Amax = 270 nm.
- 286 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 50. Synthesis of N4,2'-0-dimethyl Cytidine (00901074004):
Scheme 43
0
NH NH 1,2,4-
triazole
0 POCI3, 00
HO TBDMSCI0 ____________________________________
TBDMS-0
imidazole Et3N, MeCN
OH OCH3 0 OCH3
TBDMS-
130 151
,CH3
N2 HN
N
N Me2
TBDMS-0 0 40% aq. solution TBDMS-0
MeCN
TBDMS-0 OCH3 TBDMS-0 OCH3
153
152 NHCH3
N
0
HO
O
TBAF
THF OH OCH3
154
Synthesis of Compound 151. To a solution of compound 130 (5.16 g, 20.0 mmol)
in dry DMF (50 mL)
were added tert-butyldimethylsilyl chloride (12.0 g, 80 mmol) and imidazole
(6.8 g, 100.0 mmol). The
clear reaction mixture was stirred at room temperature for 24 h. Water was
added, and the mixture
was extracted with ethyl acetate. The combined organic phase was washed with
brine, and dried over
anhydrous Na2504. The drying agent was filtered off, and the filtrate was
concentrated to dryness
under reduced pressure. The crude product thus obtained was purified by flash
chromatography on a
silica gel column using petroleum ether-ethyl acetate (5:1 to 1:1) to give 8.3
g compound 151 as
colorless oil in 85%.
Synthesis of Compound 152. To a stirred mixture of 1,2,4-triazole (2.24 g,
32.5 mmol) in anhydrous
methylene chloride (20 mL) at 0 C was added POCI3 (1.04 g, 6.8 mmol) slowly.
Triethylamine (3.09
g, 30.6 mmol) was then added dropwise. The resulted suspension was stirred for
30 min. A solution
of compound 151 (1.7 g, 3.4 mmol) in anhydrous dichloromethane (5 mL) was
added. The reaction
mixture was then continuously stirred overnight and quenched with water. The
mixture was extracted
with dichloromethane. The combined organic phase was washed with brine and
dried over anhydrous
Na2504. The drying agent was filtered off, and the filtrate was concentrated
under reduced pressure
to give 1.9 g crude product compound 152 which was carried to the next step
without further
purification.
- 287 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Synthesis of Compound 153. To a stirred solution of compound 152 (1.9 g, crude
obtained above) in
absolute ethanol (20 mL) was added methylamine MeNH2 (20 mL, 40% aq.
solution). The reaction
mixture was stirred at room temperature for 30 min. The reaction mixture was
concentrated under
reduced pressure to dryness. The crude product was purified by flash
chromatography on a silica gel
column using petroleum ether-ethyl acetate (5:1 to 1:1) resulting in 1.5 g of
compound 153 (86%) as
a white solid.
Synthesis of N4,2'-0-Dimethylcytidine (154). Tetrabutylammonium fluoride
trihydrate (1.58 g, 6.0
mmol) was added to a stirred solution of compound 153 (1.5 g, 3.0 mmol) in dry
THF (15 mL), and
the reaction mixture was stirred at room temperature for 12 h. The mixture was
then concentrated
under reduced pressure. The crude product was purified by flash chromatography
on a silica gel
column using methylene chloride-methanol (20:1) to give final product compound
154 (500 mg,
61.4%) as a white solid. HPLC purity: 97.56%. 1H NMR (DM50-d6): 6 7.81 (d, 1H,
J = 7.6Hz), 7.68
(m, 1H, NH), 5.86 (d, 1H, J= 4.4Hz), 5.72 (d, 1H, J= 7.2Hz), 5.07 (m, 2H, J=
8.0Hz), 4.05 (s, 1H),
3.81 (t, 1H, J= 2.8Hz), 3.60-3.70 (m, 2H), 3.53-3.58 (m, 1H), 3.36 (d, 3H, J=
4.8Hz), 2.74 (d, 3H, J=
4.8Hz). ESI MS, m/e 272 (M + H)+, 273 (2M + H) . UV, Amax= 270.50 nm, =
11557 L.mor1.cm-1, y=
11557 x, R2 = 0.9991 (C = 2.7368 x 10-5-8.2103 x 10-5mol/L).
- 288 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 51. Synthesis of 5-carbamoylmethyl uridine (03601013036):
Scheme 44
0
0 0 II Br
HN
HN HN J=
Ac0 0 N
HO OjN Ac20 Ac0 ONBr2, Ac20
0
Py AcOH
98% 60% Ac0 OAc
OH OH Ac0 OAc 156
146 155
0
,
o o 0\
o 00 0
)cBr
Ph N 0\ Ph N 0
DMAP, Et3N 0 _________________________ 0
_____________________________ Ac0 ON Ac0 0 N
Bz-CI
64% DBU, THF
50%
Ac0 OAc Ac0 OAc
157 158
0
0
OCH3
HN NH2
Na0Me HN
0 NH4OH
, HO 0 N 0
0' HO 0 N
Me0H, heat rt o/n
Dowex-50 (H )
OH OH
OH OH
150 160
Synthesis of 2',3',5'-tri-O-acetyluridine (155). To a solution of uridine 146
(1.0 g, 4.0 mmol) in 20 mL
of pyridine was added 2 mL (2.16 g, 21.0 mmol) of acetic anhydride. The
resulting reaction mixture
was heated to 60 C for 3 h, and the TLC indicated its completion. The
reaction mixture was
concentrated, and the residue was purified by flash chromatography on a silica
gel column using
dichloromethane-methanol (80:1) as eluent giving 1.2 g desired product 155 in
79% yield.
Synthesis of 5-bromo-2',3',5'-tri-O-acetyluridine (156). Compound 155 (1.2 g,
3.0 mmol) was
dissolved in 20 mL of acetic acid, and 1.2 mL (1.25 g, 11 mmol) acetic
anhydride was added. The
resulting mixture was cooled to 0 C in an ice bath, and bromine (0.7 g, 4.0
mmol) was added slowly
under stirring. The reaction flask was sealed, and the mixture was stirred at
room temperature
overnight. Ethanol was added slowly, and the mixture was concentrated under
reduced pressure to
dryness. The residue was co-evaporated with ethanol and purified by flash
chromatography on a
silica gel column using methylene chloride-methanol (80:1) as eluent providing
1.3 g desired bromo
product 156 in 89% yield. 1H NMR (CDCI3) 6 9.10 (br, 1H), 7.82 (s, 1H), 6.07
(m, 1H), 5.26-5.35 (m,
2H), 4.30-4.41(m, 3H), 2.20 (s, 3H), 2.11 (s, 3H), 2.09 (s, 3H).
- 289 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Synthesis of 5-bromo-N3-benzoy1-2',3',5'-tri-O-acetyluridine (157). Compound
156 (1.3 g, 2.9 mmol)
was dissolved in 40 mL of dichloromethane, and it was cooled to 0 C. To the
stirred solution were
added N,N-dimethylaminopyridine (DMAP) (0.50 g, 4.0 mmol) and triethylamine
(0.41 mL, 0.303 g,
3.0 mmol). Benzoyl chloride (0.70 mL, 0.83 g, 5.79 mmol) was then added
slowly. The reaction
mixture was stirred at room temperature for 30 minutes, and treated with a
mixture of pyridine and
water. It was then extracted with dichloromethane. The organic phase was
washed with water and
dried over anhydrous sodium sulfate. The drying agent was filtered off, and
the filtrate was
concentrated under reduced pressure. The crude product was purified by flash
chromatography on a
silica gel column using methylene chloride-methanol (80:1) as eluent providing
1.4 g of desired
product 157 as white foam in 87% yield.
Synthesis of N3-benzoy1-2',3',5-0-triacetyluridine-5-malonic acid dimethyl
ester (compound 158). N3-
Benzoy1-5-bromo-2',3',5'-tri-O-acetyluridine (157) (1.40 g, 2.53 mmol) was
dissolved in anhydrous
THF (20-30 mL). To this solution were added dimethyl malonate (320 uL, 2.8
mmol) and DBU (450
uL). The reaction mixture was stirred at room temperature overnight, and small
amount of acetic acid
was added to quench the reaction. The mixture was concentrated and the residue
was purified by
flash chromatography on a silica gel column using dichloromethane-methanol
(80:1) as eluent
providing 1.30 g desired product 158 as white foam in 84% yield.
Synthesis of 5-(methoxycarbonyl)methyluridine (uridine 5-accetic acid methyl
ester) (159). To a
solution of N3-benzoy1-2',3',5-tri-O-acetoxyuridine-5-malonic acid dimethyl
ester (158) (1.30 g, 2.1
mmol) in 100 mL of absolute methanol was added sodium methoxide (25% in
methanol, 3.5 mL). The
reaction mixture was stirred at 50 C for 16 h, and diluted with methanol.
Sodium bicarbonate was
added to the mixture, and the solid was filtered. The filtrate was
concentrated under reduced
pressure. The residue was purified by flash chromatography on a silica gel
column using
dichloromethane-methanol (20:1) as eluent providing 400 mg desired product 159
as white foam in
about 70% yield. 1H NMR (DM50-d6): 6 11.46 (d, 1H, J= 3.0 Hz), 7.56 (d, 1H, J=
3.6 Hz), 4.91 (d,
1H, J = 3.6 Hz), 4.79 (t, 1H, J = 4.2Hz), 4.70 (d, 1H, J = 4.2Hz), 4.49 (d,
1H, J = 3.0Hz), 3.82-3.88 (m,
2H), 3.66-3.67 (m, 1H), 3.57-3.61 (m, 1H), 3.40-3.47 (m, 1H), 3.09 (s, 3H).
ESI mass spectrum m/z
339 (M + Na).
Synthesis of Compound 160. A mixture of compound 159 (1.0 g) in ammonia
saturated methanol
solution (40 mL) was stirred for 2 days. Upon completion of the reaction as
monitored by TLC, the
reaction mixture was concentrated under reduced pressure to dryness. The crude
product thus
obtained was recrystallized from methanol giving the desired compound 160 with
95% HPLC purity. It
was characterized by NMR, MS and UV spectral analyses. 1H NMR (D20): 6 7.77
(s, 1H), 5.82 (d, 1H,
J= 4.0 Hz), 4.22-4.28 (m, 1H), 4.11-4.20 (m, 1H), 3.95-4.05 (m, 1H), 3.60-3.80
(m, 1H), 3.20-3.30 (m,
2H). ESI mass spectrum m/z 302 (M + H)+, 324 (M + Na), 625 (2M + Na). UV, 2max
= 260 nm.
- 290 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 52. Synthesis of 5-(isopentenylamino(TFA)methyl)uridine (03601013044):
Scheme 45
0 0 0
ANH)cz
HN
HN
formaldehyde Mel
HO 0 N HO
N3
_____________________________ HO ON
pyrrolidine
OH OH OH OH OH OH
146 161 162
0 OH 3 0 OH3
CH3 H N, N k,n3 HN N 1/4_,E13
H
H2N,
L4-13 HO ce"--N-"" HO 0 N
163
OH OH OH OH
164 165
Synthesis of Compound 161. A mixture of compound 146 (6.0 g, 24.6 mmol) and
formaldehyde
(12.28 g, 123 mmol, 30% aq. solution, 5 eq) was diluted with water (12 mL).
The resulting reaction
mixture was cooled to 10 C, and pyrrolidine (10.5 g 147 mmol, 6 eq) was
added. The reaction
mixture was stirred at 100 C for 2 h. Upon completion of the reaction as
monitored by TLC, the
reaction mixture was concentrated under reduced pressure to dryness. The crude
product was
purified by flash chromatography on a silica gel column using methylene
chloride-methanol (7:1 to
5:1) containing 0.2% ammonium hydroxide, resulting in compound F as white
foam. This crude
product thus obtained was recrystallized from isopropanol giving the desired
compound 161 as a
white solid with 97% HPLC purity.
Synthesis of Compound 162. To a stirred solution of compound 161 (3.0 g, 9
mmol) in absolute
methanol (50 mL) was added methyl iodide (24 g). The reaction mixture was
stirred at room
temperature for 3 days. Upon completion of the reaction as monitored by TLC,
the reaction mixture
was concentrated under reduced pressure giving crude product compound 162
which was carried to
the next step without further purification.
Synthesis of Compound 164. To a stirred solution of compound 162 (crude
obtained above) in
absolute methanol (45 mL) was added 1-bromo-3-methyl-2-butene 163 (5.4 g). The
reaction mixture
was stirred at room temperature for 1 h, and concentrated under reduced
pressure to dryness. The
crude product was purified by flash chromatography on a silica gel column
using methylene chloride-
methanol (7:1 to 5:1 to 4:1) resulting in 2.9 g compound 164.
Synthesis of Compound 165. To a solution of compound 164 (2.9 g 8.5 mmol) in
dry pyridine (50 mL)
was added trifluoroacetic anhydride (5 mL, 35.4 mmol, 4 eq). The reaction
mixture was stirred at
room temperature for 3 days as monitored by TLC for its completion. The
reaction mixture was
- 291 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
concentrated under reduced pressure to dryness. The crude product was purified
by flash
chromatography on a silica gel column using methylene chloride-methanol (25:1
to 15:1 with 0.2%
ammonium hydroxide) giving final product compound 165 (410 mg, 10.2%) as a
white solid. HPLC
purity: 98%. The product was characterized by NMR, MS and UV spectral
analyses. 1H NMR (DMS0-
d6 400Hz): 6 11.50 (d,1H,NH), 7.55 (d, 1H, J= 10.4Hz), 5.98-6.02 (m, 1H), 5.44-
5.59 (m, 2H), 5.08 (s,
1H), 4.94 (t, 1H, J= 5.2Hz), 3.91-4.22 (m, 6H), 3.75 (t, 1H, J= 5.2Hz), 3.52-
3.61 (m, 2H), 1.58-1.70
(m, 6H). ES1 MS, m/e 438 (M + H)+, 460 (M + Na), 897 (2M + Na). UV, Amax= 275
nm.
Example 53. Synthesis of 5-{lsopentenylamino(TFA)methy1}2-thiouridine
(03601013043):
Scheme 46
0 0 CH3
HN CI CH3
HN CH3
H
SN H NCH23
HO HO SN
(CF300)20
00 020
142 166
0 CH3 0 CH3
)c HN N 1/4_,n3 HN N L,F13
O
HO s N _CE
3
HO S N 0 CF3
acid
C 0 OH OH
X
168
167
Synthesis of Compound 166. To a stirred solution of compound 142 (crude) in
dioxane (50 mL) was
added excess amount of 1-amino-3-methyl-2-butene. The reaction mixture was
stirred at room
temperature overnight. Upon completion of the reaction as monitored by TLC,
the reaction mixture
was concentrated under reduced pressure to dryness. The crude product was
purified by flash
chromatography on a silica gel column using ethyl acetate - petroleum ether
(1:3 to 1:1) resulting in
compound 166 (3.1 g) which was used for next step without further
purification.
Synthesis of Compound 167. To a stirred solution of compound 166 (3.1 g, 7
mmol) in dry pyridine
(50 mL) was cooled to 0 C, and trifluoroacetic anhydride (12 mL, 18 g, 8
mmol, 1.2 eq) was added
under N2 atmosphere. The reaction mixture was stirred at room temperature for
1 h. Upon completion
of the reaction as monitored by TLC, the reaction mixture was treated with
methylene chloride (100
mL) and sodium bicarbonate solution (100 mL, 5%). The organic phase was
separated, and the
aqueous phase was extracted with dichloromethane. The combined organic phase
was dried over
anhydrous Na2504. The drying agent was filtered off, and the filtrate was
concentrated under reduced
- 292 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
pressure to dryness. The crude product was purified by flash chromatography on
a silica gel column
using ethyl acetate - petroleum ether (1:5 to 1:3) resulting in desired
compound 167 which was used
for next step without further purification.
Synthesis of Compound 168. A mixture of compound 167 (1 g) and hydrochloric
acid (1 M, 20 mL)
was stirred at room temperature for 30 min. Sodium carbonate was added to
neutralize the reaction
mixture. The solid material was filtered off, and the filtrate was
concentrated under reduced pressure
to dryness. The crude product was purified by flash chromatography on a silica
gel column giving the
desired compound 168 (370 mg) with 98.27% HPLC purity. Compound 168 was
characterized by
HNMR, MS and UV spectral analyses. 1H NMR (DMSO-d6400Hz): 6 12.78 (d, 1H, NH),
8.10 (d, 1H, J
= 23.2Hz), 6.53-6.56 (m, 1H), 5.42 (d, 1H, J= 5.6Hz), 5.06-5.16 (m, 3H), 3.90-
4.23 (m, 7H), 3.59-3.68
(m, 2H), 1.56-1.69 (m, 6H). ESI MS, m/e 454 (M + H)+, 476 (M + Na). UV, Amax=
277 nm.
Example 54. Synthesis of 5-{lsopentenylamino(TFA)methyl}-T-0-methyluridine
(03601073043):
Scheme 47
HNN )c7
ANH
Nj
Mel HN
j
formaldehyde HO ______________ O HO ON
HON 0 _______________________
pyrrolidine
OH OCH3 OH OCH3
OH OCH3
169 170
130
0 CH3
0 CH3
CH3 H3
CH3
H2NCH3 HN
(CF3C0)20 HO 0 N 0 CF3
O
OH OCH3 H OCH3
172
171
Synthesis of Compound 169. To a mixture of compound 130 (10.32 g, 40.0 mmol)
and water (20 mL)
were added pyrrolidine (14.2 g, 200.0 mmol) and paraformaldehyde (13.8 mL,
200.0 mmol). The
reaction mixture was stirred at 105 C for 48 h and concentrated under reduced
pressure. The crude
product was purified by silica gel chromatography (MeOH:DCM=1:15) on a silica
gel column giving
compound 169 (4.3 g, 32%) as oil.
Synthesis of Compound 170. Compound 169 (4.3 g, 12.6 mmol) was dissolved in
Me0H (50 mL),
and Mel (7.8 mL, 126.0 mL) was added. The reaction mixture was stirred at room
temperature for 12
h, and then concentrated providing crude compound 170 which was used for next
step without further
purification.
- 293 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Synthesis of Compound 171. The crude compound 170 (obtained above) was
dissolved in Me0H (40
mL), and to the stirred solution was added compound D (3.2 g, 37.8 mmol). The
reaction mixture was
stirred at room temperature for 72 h and concentrated under the reduced
pressure. The crude product
was purified by silica gel chromatography (MeOH:DCM=1:40) giving compound 171
(1.0 g, 22%) as a
white solid.
Synthesis of Compound 172. Compound 171 (1.0 g, 2.8 mmol) was dissolved in
anhydrous pyridine (10
mL), and the solution was cooled to 0 C. The trifluoroacetic anhydride (2.3
g, 11.2 mmol) was added,
and the reaction mixture was stirred at room temperature for 72 h. The
solution was then concentrated,
and the residue was purified by silica gel chromatography (EA:PE=3:2) on a
silica gel column resulting in
the desired compound 172 (0.33 g, 25%) as a white foam with 99.5% HPLC purity.
It was characterized
by NMR, MS and UV spectral analyses. 1H NMR (CDCI3, 400Hz): 6 8.94 (s, 1H),
8.34 (s, 1H), 5.96-5.97
(d, 1H, J= 3.6Hz), 5.06-5.09 (t, 1H, J= 6.8Hz), 4.29-4.41 (m, 1H), 3.78-4.23
(m, 8H), 3.56-3.60 (d, 3H, J
= 15Hz), 3.35-3.82 (m, 1H), 2.75-2.77 (d, 1H, J= 6.4Hz), 1.63-1.74 (t, 6H, J=
12.8Hz). ESI MS, m/e 452
(M + H)+, 474 (M + Na). UV, Amax= 267nm.
Example 55. Synthesis of N2,2'-0-dimethylguanosine (00901072014):
Scheme 48
N"---)LNH HS = N---)LNH
I
Ac20
N N NH2 _____________ I p-thiocresol NN NH __s
N Ac0 40
DMAP, Et3N AcO0
_______________________________ N NH2
formaldehyde, AcOH
rT Et0H, low yield
OH OCH3 OAc OCH3
OAc OCH3
173 174 175
0 0
I N---)LNH
I
NaBH4 NN NHCH3 N NHCH3
Ac0 AcOH
DMSO HO
OAc OCH3 OH OCH3
176 177
Synthesis of Compound 174: To a stirred solution of 2'-0-methylguanosine
(compound 173, 3.0 g, 10.0
mmol) in anhydrous pyridine was added acetic anhydride (5.0 mL) at 0 C. The
resulted reaction mixture
was stirred at room temperature for 4 h. Ethanol (5.0 mL) was added, and the
mixture was concentrated
under reduced pressure. The residue was purified by flash chromatography on a
silica gel column giving
3.0 g compound 174 as light yellow solid.
Synthesis of Compound 175: To a stirred solution of compound 174 (3.0 g, 7.8
mmol) in 60 mL of ethanol
were added p-thiocresol (3.0 g, 24 mmol), 37% aqueous formaldehyde (1.0 ml, 24
mmol), and acetic acid
(6 ml), and the resulted reaction mixture was refluxed for 4-6 hr as monitored
by TLC. The reaction
mixture was cooled, and the resulting colorless precipitate was collected by
filtration giving 2.5 g
compound 175 as light yellow solid.
- 294 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Synthesis of Compound 177: Sodium borohydride (0.7 g, 18.0 mmol) was added to
a solution of
compound 175 (3.0 g, 5.8 mmol) in dimethyl sulfoxide (15 mL). The reaction
mixture was heated at 100
C for 1-2 hr, and then cooled to room temperature. It was then quenched with
acetic acid/ethanol (50
mL, v: v = 1:10). The resulted colorless precipitate was filtrated out, and
washed thoroughly with
methanol. The crude product was dried under reduced pressure, and water was
added. After further
evaporation of water, the residue was crystallized from water to give N2, 2'-
dimethylguanosine 177 as a
white solid (0.62 g, 43%). HPLC purity: 98 `)/0, ESI mass spectrum m/z 312.8
[M + Hr , 623 [2M + 1], 1H
NMR (300MHz, DMSO-d6) 6 11.85 (br, 1H), 7.93 (s, 1H), 7.60( br, 1H), 5.84 (d,
J= 3.9Hz, 1H), 4.82-
5.10 (br, 2H), 4.26-4.29 (m, 2H), 3.90 (s, 1H), 3.53-3.57 (m, 2H), 3.35(s,
3H), 2.78 (s, 3H). ESI mass
spectrum m/z 312 (M + H)+, 623 (2M + H)+. UV, Lmax= 258 nm.
Example 56. Synthesis of 5-methoxycarbonylmethy1-2-thiouridine (03601013035):
Scheme 49
NH2
OMe Na0Me OMe Na
_______________________________________ Me0 0CH3 HN
OCH3
--y0Me
HCOOEt H OMe Me0H 0
0 heat, S N
0 0 0 179 Na0 - then AcOH,
H20
178 5-
methyoxycarbonyl 181
180methyl alpha-formyl methyl 2-
thiouracil
dimethyl succinate succinate
0 0
j
1) HMDS, heat Hy )H-r-
OCH3 0
Na0Me, Me0H HN
OCH3
0
Ac0 Ac0 S N HO ______________________ S N
c,-0,7,0Ac 0
control
2)
Ac0 OAc Ac0 OAc OH OH
115 182 183
Synthesis of 5-Methoxycarbonylmethy1-2-thiouracil (181): A mixture of sodium
methoxide (13.5 g, 0.25
mol) in 200 mL of diethyl ether was cooled to 0 C, and it was added slowly to
a stirred mixture of
dimethyl succinate 178 (36.5 g, 0.25 mol) and ethyl formate (18.5 g, 0.25
mol). The reaction mixture was
stirred at 0 C for 3 h, and at room temperature overnight. The solvent was
evaporated, and the residue
was washed thoroughly with petroleum ether resulting intermediate 180. The
crude intermediate 180 was
dissolved in methanol, and 19 g (0.25 mol) of thiourea was added. The reaction
mixture was refluxed
overnight. It was filtered, and the solid was washed with methanol. The
filtrate was concentrated under
reduced pressure. Flash chromatographic purification on a silica gel column
resulting in the desired
product 181 in 20% yield.
Synthesis of Glycosylated Compound 182: A mixture of 5-methoxycarbonyl methyl-
2-thiouracil 181 (2.0 g,
10 mmol), 50 mL of HMDS, and catalytic amount of ammonium sulfate (50 mg) was
refluxed at 130 C.
After the mixture became clear solution, excess amount of HMDS was evaporated
under reduced
pressure. The residue was dissolved in 30 mL of 1,2-dichloromethane. To this
solution was added
protected riboside 115 (10.5 g), followed by addition of 1.73 mL (15 mmol) of
SnC14. The reaction mixture
was stirred at room temperature for 1 h, and treated with dichloromethane and
saturated sodium
- 295 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
bicarbonate. The organic phase was separated, and the aqueous phase was
extracted with
dichloromethne. The combined organic phase was extracted with dichloromethane.
The combined
organic phase was dried over anhydrous sodium sulfate. After concentrated
under reduced pressure, the
crude product was purified giving desired product 182.
Synthesis of Compound 183: 3 mL of sodium methoxide solution in methanol was
added to a solution of
compound 182 (1.2 g) in 100 mL of anhydrous methanol. The reaction mixture was
stirred at room
temperature for 1 h till solid disappeared. The reaction mixture was adjusted
to week acid with diluted
hydrochloric acid. It was then neutralized with sodium bicarbonate. The
solvent was concentrated, and
the residue was purified by flash chromatography on a silica gel column
providing final product 182 with
99% HPLC purity. 1H NMR (400MHz, DMS0d6) 6 12.73 (s, 1H), 8.17 (s, 1H), 6.54-
6.55 (d, 1H), 5.44-5.45
(d, 1H), 5.24-5.25 (d, 1H), 5.10-5.12 (d, 1H), 3.90-4.04 (m, 3H), 3.72-3.80
(m, 1H), 3.61(s, 4H), 3.29 (s,
3H); Mass Spectrum: m/z 332.9.0 (M)+, 333.8 (M + H)+, 354.9 (M + Na - H)+,
686.7 (2M + Na - H) . UV,
2max = 260 nm.
Example 57. Synthesis of 5-methyl-N-TFA-aminomethy1-2-selenouridine
(03601013048):
Scheme 50
.0
HN NH3 HN N -CH3 N
-CH3
HO S N 0CF3 TBDMSCI TBDMSO SN 0CF3 Mel,DBU
TBDMSO -, cj/ -CF3
1:)4
imidazole DMF
r-r
DMF
OH OH TBDMSO OTBDMS TBDMSO OTBDMS
145 184 185
0
0
HNN-CH3
1)NaBH4,Se, Hy-kr, y-CH3
ethanol HO SeN 0 CF3
TBDMSO SeN 0r'CF3 TBAF
2)ethanol,
rt,3 d
r-r THF
TBDMSO OTBDMS OH OH
1
186 87
Synthesis of Compound 184: A mixture of compound 145 (3.80 g, 9.52 mmol), t-
butyldimethylsilyl chloride
(14.35 g, 95.2 mmol), imidazole (7.54 g, 114.24 mmol) in 20 ml of anhydrous
DMF was stirred at 60 C
for 12 h. The solvent was concentrated under reduced pressure, and the residue
was treated with water
and ethyl acetate. The organic phase was separated and the aqueous phase was
extracted with ethyl
acetate. The organic phase was dried over anhydrous sodium sulfate. The drying
agent was filtered off,
and the filtrate was concentrated. The residue was purified by flash
chromatography on a silica gel
column providing 6.3 g product 184.
Synthesis of compound 185: Methyl iodide (9.56 g, 70.0 mmol, 10 eq) was added
to the well stirred
mixture of compound 184 (5.20 g, 7.0 mmol) and sodium bicarbonate (0.85 g,
10.12 mmol, 1.45 eq) in
anhydrous DMF. The resulting reaction mixture was stirred at room temperature
for 8-9 h, as indicated for
- 296 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
the completion of the reaction by TLC. The reaction mixture was treated with
dichloromethane and water.
The organic phase was separated, and the aqueous phase was extracted with
ethyl acetate. The organic
phase was dried, and the solvent was concentrated. The residue was purified by
flash chromatography
on a silica gel column providing 5.60 g desired product 185.
Synthesis of compound 186: Compound 185 (5.0 g, 6.61 mmol) was dissolved in 20
mL of anhydrous
ethanol. Sodium borohydride (1.30 g, 33.0 mmol) and metal selenium (2.10 g,
26.4 mmol, 8 eq) in a
separate round bottom flask was cooled to 0 C. Under nitrogen protection and
protection from light, 20
mL of anhydrous ethanol was added slowly. The reaction mixture was stirred for
30 min at 0 C till the
reaction mixture became clear orange solution. The solution of compound 185 in
ethanol prepared above
was added to the freshly prepared NaSeH4 solution. The reaction mixture was
stirred at room
temperature for 2 days, and treated with dichloromethane and water. The
organic phase was separated,
and the aqueous phase was extracted with dichloromethane. The organic phase
was concentrated, and
the residue was purified by flash chromatography on a silica gel column
providing 5.0 g of desired product
186.
Synthesis of compound 187: To a stirred solution of compound 186 (5.0 g, 6.34
mmol) in 20 mL of THF
was added 38 mL tetrabutyl ammonium fluoride. The reaction mixture was stirred
at room temperature for
2 h, and concentrated directly under reduced pressure. The residue was
purified by flash chromatography
on silica gel column. It was purified four times by column providing 120 mg of
the desired final product
187 with 95% HPLC purity. MS ES, M/z 447 (M + H)+, 469.8 (M + Na). UV, Amax=
315nm.
Example 58. Synthesis of 5-methyl dihydrouridine (03601013039):
Scheme 51
0 0
)c/
HN)C FIN
1 Rh/A103
HO 0N HO ON
hydrogenation
water
OH OH OH OH
188 189
5-Methyl-5,6-dihydrouridine 189: To a solution of 5-methyluridine 188 (3.0 g)
in water (500 mL) was
added catalyst 5% Rh/C (936 mg). The mixture was shaken in an atmosphere of
hydrogen (-0.34 MPa)
at room temperature for 12 h. The catalyst was filtered off, and the filtrate
was concentrated under
reduced pressure to dryness. Several recrystallization processes using
ethanol/ethyl acetate solvent
system yielded a mixture of stereoisomeric product 189 (2.5 g, 82%) with 99%
HPLC purity, two isomers
in total. Then further recrystallization from methanol/ether resulted in one
isomer-enriched sample (150
mg) as indicated by NMR because HPLC could not separate these two peaks. 1H
NMR (400MHz,
DMS0d6) 6 10.20 (s, 1H), 5.60-5.70 (m, 1H), 5.02-5.10 (m, 1H), 4.88-4.93 (m,
1H), 4.75-4.85 (m, 1H),
3.89-4.02 (m, 1H), 3.82-3.88 (m, 1H), 3.63-3.70 (m, 1H), 3.32-3.55 (m, 3H),
2.95-3.10 (m, 1H), 2.52-2.65
(m, 1H); Mass Spectrum: m/z 261 (M + H)+, 283 (M + Na) . UV, Amax = 220 nm.
- 297 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 59. DNA and mRNA sequences for constructs used to screen compounds of
the
invention
SEQ ID NO:5 GCSF DNA
GGGAGATCAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACCATGGCCGGTCCCGCGA
CCCAAAGCCCCATGAAACTTATGGCCCTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGT
CCAAGAAGCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTGAAGTGTCTG
GAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAGAGAAGCTCTGCGCGACATAC
AAACTTTGCCATCCCGAGGAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCT
CTCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTCCCAGCTCCACTCCGGTT
TGTTCTTGTATCAGG GACTGCTGCAAG CCCTTGAG GGAATCTCGCCAGAATTG GGCCCGACG CT
GGACACGTTGCAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATGGAGGAACT
GGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGCAATGCCGGCCTTTGCGTCCGCGTTTC
AG CGCAGG GCG GGTGGAGTCCTCGTAGCGAG CCACCTTCAATCATTTTTG GAAGTCTCGTACC
GGGTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGGAGCCTCGGTGGCCATGCTTCTTG
CCCCTTGGGCCTCCCCCCAGCCCCTCCTCCCCTTCCTGCACCCGTACCCCCGTGGTCTTTGAAT
AAAGTCTGAGTGGGCGGCTCTAGA
SEQ ID NO:6 GCSF mRNA
GGGAGAUCAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCCACCAUGGCCGGUCCCGCG
ACCCAAAGCCCCAUGAAACU UAUGGCCCUGCAG U UGCUGCU U UGGCACUCGGCCCUCUGGAC
AG UCCAAGAAGCGACUCCUCUCGGACCUGCCUCAUCG U UGCCGCAGUCAU UCCU U U UGAAGU
GUCUGGAGCAGG UGCGAAAGAU UCAGGGCGAUGGAGCCGCACUCCAAGAGAAGCUCUGCGC
GACAUACAAACU U UGCCAUCCCGAGGAGCUCG UACUGCUCGGGCACAGCU UGGGGAU UCCCU
GGGCUCCUCUCUCG UCCUGUCCG UCGCAGGCU U UGCAGU UGGCAGGG UGCCU U UCCCAGCU
CCACUCCGG U U UGU UCU UGUAUCAGGGACUGCUGCAAGCCCU UGAGGGAAUCUCGCCAGAA
U UGGGCCCGACGCUGGACACGU UGCAGCUCGACGUGGCGGAU U UCGCAACAACCAUCUGGC
AGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUGCAGCCCACGCAGGGGGCAAUGCCGGC
CU U UGCG UCCGCG U U UCAGCGCAGGGCGGG UGGAGUCCUCG UAGCGAGCCACCU UCAAUCA
UUUU UGGAAG UCUCGUACCGGG UGCUGAGACAUCU UGCGCAGCCG UGAUAAUAGGCUGGAG
CCUCGG UGGCCAUGCU UCU UGCCCCU UGGGCCUCCCCCCAGCCCCUCCUCCCCU UCCUGCA
CCCGUACCCCCG UGG UCU U UGAAUAAAGUCUGAGUGGGCGGCUCUAGA
SEQ ID NO:7 Luciferase DNA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACCATGGAAGATGCGAAGAA
CATCAAGAAGGGACCTGCCCCGTTTTACCCTTTGGAGGACGGTACAGCAGGAGAACAGCTCCA
CAAGGCGATGAAACGCTACGCCCTGGTCCCCGGAACGATTGCGTTTACCGATGCACATATTGAG
GTAGACATCACATACGCAGAATACTTCGAAATGTCGGTGAGGCTGGCGGAAGCGATGAAGAGAT
ATGGTCTTAACACTAATCACCGCATCGTGGTGTGTTCGGAGAACTCATTGCAGTTTTTCATGCCG
GTCCTTGGAGCACTTTTCATCGGGGTCGCAGTCGCGCCAGCGAACGACATCTACAATGAGCGG
- 298 -
SUBSTITUTE SHEET (RULE 26)

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
GAACTCTTGAATAGCATGGGAATCTCCCAGCCGACGGTCGTGTTTGTCTCCAAAAAGGGGCTGC
AGAAAATCCTCAACGTGCAGAAGAAGCTCCCCATTATTCAAAAGATCATCATTATGGATAGCAAG
ACAGATTACCAAGGGTTCCAGTCGATGTATACCTTTGTGACATCGCATTTGCCGCCAGGGTTTAA
CGAGTATGACTTCGTCCCCGAGTCATTTGACAGAGATAAAACCATCGCGCTGATTATGAATTCCT
CGGGTAGCACCGGTTTGCCAAAGGGGGTGGCGTTGCCCCACCGCACTGCTTGTGTGCGGTTCT
CGCACGCTAGG GATCCTATCTTTG GTAATCAGATCATTCCCGACACAG CAATCCTGTCCGTG GT
ACCTTTTCATCACGGTTTTGGCATGTTCACGACTCTCGGCTATTTGATTTGCGGTTTCAGGGTCG
TACTTATGTATCGGTTCGAGGAAGAACTGTTTTTGAGATCCTTGCAAGATTACAAGATCCAGTCG
GCCCTCCTTGTGCCAACGCTTTTCTCATTCTTTGCGAAATCGACACTTATTGATAAGTATGACCTT
TCCAATCTGCATGAGATTGCCTCAGGGGGAGCGCCGCTTAGCAAGGAAGTCGGGGAGGCAGTG
GCCAAGCGCTTCCACCTTCCCGGAATTCGGCAGGGATACGGGCTCACGGAGACAACATCCGCG
ATCCTTATCACGCCCGAGGGTGACGATAAGCCGGGAGCCGTCGGAAAAGTGGTCCCCTTCTTT
GAAGCCAAGGTCGTAGACCTCGACACGGGAAAAACCCTCGGAGTGAACCAGAGGGGCGAGCTC
TGCGTGAGAGGGCCGATGATCATGTCAGGTTACGTGAATAACCCTGAAGCGACGAATGCGCTG
ATCGACAAGGATGGGTGGTTGCATTCGGGAGACATTGCCTATTGGGATGAGGATGAGCACTTCT
TTATCGTAGATCGACTTAAGAGCTTGATCAAATACAAAGGCTATCAGGTAGCGCCTGCCGAGCTC
GAGTCAATCCTGCTCCAGCACCCCAACATTTTCGACGCCGGAGTGGCCGGGTTGCCCGATGAC
GACGCGGGTGAGCTGCCAGCGGCCGTGGTAGTCCTCGAACATGGGAAAACAATGACCGAAAAG
GAGATCGTGGACTACGTAGCATCACAAGTGACGACTGCGAAGAAACTGAGGGGAGGGGTAGTC
TTTGTGGACGAGGTCCCGAAAGGCTTGACTGGGAAGCTTGACGCTCGCAAAATCCGGGAAATC
CTGATTAAGGCAAAGAAAGGCGGGAAAATCGCTGTCTGATAATAGGCTGGAGCCTCGGTGGCC
ATGCTTCTTGCCCCTTGGGCCTCCCCCCAGCCCCTCCTCCCCTTCCTGCACCCGTACCCCCGTG
GTCTTTGAATAAAGTCTGAGTGGGCGGCTCTAGA
SEQ ID NO:8 Luciferase mRNA
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCCACCAUGGAAGAUGCGAAGA
ACAUCAAGAAGGGACCUGCCCCG U U U UACCCU U UGGAGGACGG UACAGCAGGAGAACAGCUC
CACAAGGCGAUGAAACGCUACGCCCUGGUCCCCGGAACGAU UGCGU U UACCGAUGCACAUAU
UGAGG UAGACAUCACAUACGCAGAAUACU UCGAAAUGUCGGUGAGGCUGGCGGAAGCGAUGA
AGAGAUAUGGUCU UAACACUAAUCACCGCAUCG UGGUG UGU UCGGAGAACUCAU UGCAG U U U
U UCAUGCCGGUCCU UGGAGCACU U U UCAUCGGGGUCGCAG UCGCGCCAGCGAACGACAUCU
ACAAUGAGCGGGAACUCU UGAAUAGCAUGGGAAUCUCCCAGCCGACGGUCGUG U U UGUCUCC
AAAAAGGGGCUGCAGAAAAUCCUCAACGUGCAGAAGAAGCUCCCCAU UAU UCAAAAGAUCAUC
AU UAUGGAUAGCAAGACAGAU UACCAAGGG U UCCAGUCGAUG UAUACCU U UGUGACAUCGCA
U U UGCCGCCAGGGU U UAACGAG UAUGACU UCG UCCCCGAG UCAU U UGACAGAGAUAAAACCA
UCGCGCUGAU UAUGAAU UCCUCGGG UAGCACCGGU U UGCCAAAGGGGGUGGCGU UGCCCCA
CCGCACUGCU UGUG UGCGG U UCUCGCACGCUAGGGAUCCUAUCU U UGG UAAUCAGAUCAU U
CCCGACACAGCAAUCCUG UCCGUGGUACCU U U UCAUCACGGU U U UGGCAUGU UCACGACUCU
CGGCUAU U UGAU U UGCGG U U UCAGGG UCGUACU UAUG UAUCGG U UCGAGGAAGAACUGU U U
U UGAGAUCCU UGCAAGAU UACAAGAUCCAGUCGGCCCUCCU UGUGCCAACGCU U U UCUCAU U
CU U UGCGAAAUCGACACU UAU UGAUAAG UAUGACCU U UCCAAUCUGCAUGAGAU UGCCUCAG
- 299 -
SUBSTITUTE SHEET (RULE 26)

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
GGGGAGCGCCGCU UAGCAAGGAAGUCGGGGAGGCAGUGGCCAAGCGCU UCCACCU UCCCGG
AAU UCGGCAGGGAUACGGGCUCACGGAGACAACAUCCGCGAUCCU UAUCACGCCCGAGGG U
GACGAUAAGCCGGGAGCCGUCGGAAAAG UGG UCCCCU UCU U UGAAGCCAAGG UCGUAGACC
UCGACACGGGAAAAACCCUCGGAG UGAACCAGAGGGGCGAGCUCUGCG UGAGAGGGCCGAU
GAUCAUGUCAGG U UACG UGAAUAACCCUGAAGCGACGAAUGCGCUGAUCGACAAGGAUGGG U
GG U UGCAU UCGGGAGACAU UGCCUAU UGGGAUGAGGAUGAGCACU UCU U UAUCG UAGAUCG
ACU UAAGAGCU UGAUCAAAUACAAAGGCUAUCAGG UAGCGCCUGCCGAGCUCGAG UCAAU CC
UGCUCCAGCACCCCAACAU U U UCGACGCCGGAG UGGCCGGGU UGCCCGAUGACGACGCGGG
UGAGCUGCCAGCGGCCGUGGUAGUCCUCGAACAUGGGAAAACAAUGACCGAAAAGGAGAUCG
UGGACUACG UAGCAUCACAAG UGACGACUGCGAAGAAACUGAGGGGAGGGG UAGUCU U UG U
GGACGAGGUCCCGAAAGGCU UGACUGGGAAGCU UGACGCUCGCAAAAUCCGGGAAAUCCUGA
U UAAGGCAAAGAAAGGCGGGAAAAUCGCUG UCUGAUAAUAGGCUGGAGCCUCGG UGGCCAUG
Cu UCU UGCCCCU UGGGCCUCCCCCCAGCCCCUCCUCCCCU UCCUGCACCCG UACCCCCG UG
G UCU U UGAAUAAAG UCU GAG UGGGCGGCUCUAGA
SEQ ID NO:9 EPO DNA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACCATGGGAGTGCACGAGTG
TCCCGCGTGGTTGTGGTTGCTGCTGTCGCTCTTGAGCCTCCCACTGGGACTGCCTGTGCTGGG
GGCACCACCCAGATTGATCTGCGACTCACGGGTACTTGAGAGGTACCTTCTTGAAGCCAAAGAA
GCCGAAAACATCACAACCGGATGCGCCGAGCACTGCTCCCTCAATGAGAACATTACTGTACCGG
ATACAAAGGTCAATTTCTATGCATGGAAGAGAATGGAAGTAGGACAGCAGGCCGTCGAAGTGTG
GCAGGGGCTCGCGCTTTTGTCGGAGGCGGTGTTGCGGGGTCAGGCCCTCCTCGTCAACTCATC
ACAGCCGTGGGAGCCCCTCCAACTTCATGTCGATAAAGCGGTGTCGGGGCTCCGCAGCTTGAC
GACGTTGCTTCGGGCTCTGGGCGCACAAAAGGAGGCTATTTCGCCGCCTGACGCGGCCTCCGC
GGCACCCCTCCGAACGATCACCGCGGACACGTTTAGGAAGCTTTTTAGAGTGTACAGCAATTTC
CTCCGCGGAAAGCTGAAATTGTATACTGGTGAAGCGTGTAGGACAGGGGATCGCTGATAATAGG
CTGGAGCCTCGGTGGCCATGCTTCTTGCCCCTTGGGCCTCCCCCCAGCCCCTCCTCCCCTTCC
TGCACCCGTACCCCCGTGGTCTTTGAATAAAGTCTGAGTGGGCGGCTCTAGA
35
- 300 -
SUBSTITUTE SHEET (RULE 26)

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
SEQ ID NO:10 EPO mRNA
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCCACCAUGGGAGUGCACGAGU
GUCCCGCGUGGUUGUGGUUGCUGCUGUCGCUCUUGAGCCUCCCACUGGGACUGCCUGUGC
UGGGGGCACCACCCAGAUUGAUCUGCGACUCACGGGUACUUGAGAGGUACCUUCUUGAAGC
CAAAGAAGCCGAAAACAUCACAACCGGAUGCGCCGAGCACUGCUCCCUCAAUGAGAACAUUAC
UGUACCGGAUACAAAGGUCAAUUUCUAUGCAUGGAAGAGAAUGGAAGUAGGACAGCAGGCCG
UCGAAGUGUGGCAGGGGCUCGCGCUUUUGUCGGAGGCGGUGUUGCGGGGUCAGGCCCUCC
UCGUCAACUCAUCACAGCCGUGGGAGCCCCUCCAACUUCAUGUCGAUAAAGCGGUGUCGGG
GCUCCGCAGCUUGACGACGUUGCUUCGGGCUCUGGGCGCACAAAAGGAGGCUAUUUCGCCG
CCUGACGCGGCCUCCGCGGCACCCCUCCGAACGAUCACCGCGGACACGUUUAGGAAGCUUU
UUAGAGUGUACAGCAAUUUCCUCCGCGGAAAGCUGAAAUUGUAUACUGGUGAAGCGUGUAGG
ACAGGGGAUCGCUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGCUUCUUGCCCCUUGGGCCU
CCCCCCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCCGUGGUCUUUGAAUAAAGUCUGAG
UGGGCGGCUCUAGA
-301 -
SUBSTITUTE SHEET (RULE 26)

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Example 70. In vitro transcription yields
Table 19: In vitro Transcription Yields.
Luc In Vitro EPO In Vitro
GCSF In Vitro
Chemical
Compound # Structure
Transcription Transcription Transcription
Modifications
yield(mg) yield(mg)
yield(mg)
0
00902015001 HNANH PseudoU-
(194) o o s
II H II
0 alpha-thio-TP 0.6479 0.8632 0.5522
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
00902015002
NNH 1-Methyl-
(195) o
pseudo-U- 0.6011 0.7679 0.6582
o s
II II II o
HO-P-O-P-O-P-0 alpha-thio-TP
OH OH OH
OH OH
0
-
03601015003 N ANH 1-Ethyl-
(172) o
II H H o pseudo-UTP 0.6304 1.095 0.5464
HO-P-O-P-O-P-0
OH OH OH cLO_
OH OH
0
03601015004 NANH pseudo-UTP 1-Propyl-
(173)
0.4971 0.9920 0.5976
o o o
II H H (:)
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
03601015005 F3CNANH 1-(2,2,2-
(175) o o
Trifluoroethyl) 0.4388 0.3379 0.2332
o
II H H o
HO-P-O-P-O-P-0 pseudo-UTP
OH OH OH Icç
OH OH
S
00901015006 HN ANH pseudo-UTP 2-Thio-
(193)
0.6123 1.081 0.5207
o o o
II H H (:)
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
5-
00901013002 F3C)-LNH
(352)o o o
II H H t N0 Trifluorometh 0.3662 0.3830
0.5102
HO-P-O-P-O-P-0 yl-UTP
OH OH OH Icç
OH OH
- 302 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
5-
00901014003 F3c ,..,..J,,,N
(351)o o o
II H H 1
Thl 0 Trifluorometh 0.5097 0.7886 0.7710
HO-P-O-P-O-P-0 yl-CTP
OH OH OH
OH OH
0
00901015187 HNAN 3-Methyl-
(236)
0.0152 0.0125 0.0120
o o o
pseudo-UTP
II H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
00901013004 ANH 5-Methyl-2-
(4)o o o
ii II H t N
thio-UTP 0.7580 0.8717
0.4682
HO-P-O-P-O-P-0
OH OH OH
OH OH
HN
00901014004 N N4-methyl
(346) o o o
II H H 1
N 0 CTP 1.124 1.154 0.9028
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
5-
00901014005 HON
(350)o o o
II H H 1
Thl 0 Hydroxymeth 0.4073 0.7778 0.6391
HO-P-O-P-O-P-0 yl-CTP
OH OH OH
OH OH
NH
AN
00901014006 N0
o o o 3-Methyl-CTP
0.0068 0.0060 0.0141
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
0 0
o-C))-NH UTP-5-
00901013004
(348)o o o
II H H NO
oxyacetic acid 0.6348 0.3859
0.3836
HO-P-O-P-O-P-0 Me ester
OH OH OH
OH OH
0
00901013005 Oy*NH 5-Methoxy
(358) o 0 NOO
carbonyl 0.8825 0.6432
0.6475
o o ,.L
HO-P-O-P-O-P-0 methyl-UTP
OH OH OH
OH OH
- 303 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
o
5-
00901013006 ir\JH
(353) o o o H I
N- Methylamino 0.2914 0.3060 0.3494
H0+0+0+0 methyl-UTP
OH OH OH
OH OH
0
0)-NH 5-methoxy-
00901013007 tL 0.3817 0.1727 0.1546
9 9 9 N O UTP
HO¨PI-0¨PI-0-10
OH OH OH
OH OH
0
H11)-
00901014007 N N4-Ac-CTP 0.4394 0.4351 0.3658
9 9 9 1
N 0
HO-1-0-1-0-1-0
OH OH OH Icçi
OH OH
0
NN--
00901012008 f N1-Me-GTP 0.0059 0.0032 0.0050
9 9 9 N N-NH2
HOIT'0101T'0
OH OH OH
OH OH
NH2
03601011002 NI---1,,,--N
I 2-Amino-ATP 0.1215 0.2612 0.1567
(154) N N-.- NH2
H01,0c,01,0
OH OH OH
OH OH
NH2
00901011003,N----N
Ns 1 ) 8-Aza-ATP 0.0262 0.0055 0.03
(377) 9 9 N'N'
HO-1-0-1-0-1-0
OH OH OH
OH OH
o
00901012003 N"---A, NH
I J,
(378) N't\i0
Xanthosine 0.0054 0.0032 0.0041
9 9 9-
HO¨P¨O¨P¨O¨P-0 H
OH OH OH
OH OH
NH2
03601014008 Br N
(379) 9 9 9I _L
11" -C, 5-Bromo-CTP 0.5161 0.3454 0.3685
HO-1-0-1-0-1-0
OH OH OH
OH OH
- 304 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
03601014009 N2N'¨'¨:"----'¨'1---N 5-Aminoallyl-
(381) o 0
t N CTP 0.3471 0.4943
0.4567
o
II H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
2-
03601012004
9 9 9 (N N NH. ---I - Aminopurine- 0.0690
0.0125 0.2919
(382) HO-1-0-1-0-1-0
OH OH OH 'Ic2_ riboside TP
OH OH
0
)-L NH
00901013008 tN
o o 0 S 2-Thio-UTP 0.2792
0.3630 0.3359
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
BrNH
00901013009 1
o o 0 5-Bromo-UTP 0.3352
0.2617 0.3566
II II II 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
N
00901014010 tN
o o 0 S 2-Thio-CTP 0.0073
0.0061 0.0076
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
N Alpha-thio-
00902014001
o o s tNO 0.3352 0.2669
0.2650
II II II CTP
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
H2N NL H 5-Aminoallyl-
00901013010
o o 0 0.3513 0.3732
0.4206
II II II N ICI UTP
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
)1\1H Alpha-thio-
00902013001
O o s tNLO 0.3510 0.2666 0.2605
II II II UTP
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 305 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
S
00901013011 ANN
(2) o o o I
0 4-Thio-UTP 0.1625 0.0416 0.0759
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
F3CN
0 0 0 t N
HO-P-O-P-O-P-0
II II II 5-
OH OH OH Trifluorometh
00901014003/ OH OH yl-CTP/1- 0.3405 0.4471 0.2966
00901015002 o Methyl-
Th\J)-NH pseudo-UTP
O o o o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
HO-AN
0 0 0 1
N 0 5
II II II
-
HO-P-O-P-O-P-0
OH OH OH Hydroxymeth
00901014005/ OH OH yl-CTP/1- 0.3270 0.3149 0.3705
00901015002 o Methyl-
N ANH pseudo-UTP
O o o o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
BrAN
0 0 0 1
N 0
II II II
HO-P-O-P-O-P-0
5-Bromo-
OH OH OH CTP/1-
03601014008/ OH OH 0.2594 0.3073 0.3958
00901015002 Methyl-
0
N ANH pseudo-UTP
O o o o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 306 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
F3cN
O 0 0 t N
HO-P-O-P-O-P-0
II II II 5-
OH OH OH Trifluorometh
00901014003/ OH OH yl- 0.3316 0.4486 0.4197
00901015001 o CTP/Pseudo-
HN )-NH UTP
O o o o
II II II
HO-P-O-P-O-P-0
OH OH OH k2_
OH OH
NH2
BrN
O 0 0 1
N 0
II II II
HO-P-O-P-O-P-0
OH OH OH (cL) 5-Bromo-
03601014008/ OH OH CTP/Pseudo- 0.3265 0.4879
0.2982
00901015001 o UTP
HN AN H
O 0 0 o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
F3CN
O 0 0 t N
II II II
HO-P-O-P-O-P-0
OH OH OH k:)_
OH OH
75% 5-
Trifluorometh
NH2
N yl-CTP + 25
00901014003/ t
o o o % CTP/1-
NO 0.3316 0.4008 0.4777
00901015002
ii ii ii
HO-P-O-P-O-P-0
Methyl-
OH OH OH pseudo-UTP
OH OH
0
N)-NH
O 0 0 o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 307 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
0 0 0 NO
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
Trifluorometh
NH2
00901014003/
yl-CTP + 50
N 0.3884 0.3990 0.4130 00901015002 o o o
NO % CTP/1-
H H H
HO-P-O-P-O-P-0 Methyl-
OH OH OH 'VL5 pseudo-UTP
OH OH
0
--11,NH
0 0 0
II II II
0
HO-P-O-P-O-P-0
OH OH OH '1
OH OH
NH2
F3Cõ..õA,N
t
0 0 0 N
II II II
HO-P-O-P-O-P-0
OH OH OH 2
OH OH 5 % 5-
NH2 Trifluorometh
N yl-CTP + 75
00901014003/ o o o I
NO % CTP/1- 0.3157 0.3913 0.5430
00901015002 HO-P-O-P-O-P-0
OH OH OH '1 IcL) Methyl-
OH OH pseudo-UTP
0
NANH
0 0 0
II II II
0
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 308 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
BrN
0 0 0 0
H
HO¨P¨O¨P¨O¨P-0
OH OH OH
OH OH
50% 5-
NH2 Bromo-CTP +
03601014008/ I
o o o 50 % CTP/1- 0.2897 0.4181
0.3894
00901015002 0 Methyl-
HO¨P¨O¨P¨O¨P-0
OH OH OH1cç pseudo-UTP
OH OH
0
NH
O 0
0
H H
HO¨P¨O¨P¨O¨P-0
OH OH OH
OH OH
NH2
BrN
0 0 0 0
H H H
HO¨P¨O¨P¨O¨P-0
OH OH OH
OH OH
25% 5-
NH2
N Bromo-CTP +
03601014008/ o o o I
NO 75% CTP/1- 0.3258 0.3930
0.4911
00901015002 HO¨P¨O¨P¨O¨P-0 Methyl-
OH OH OH pseudo-UTP
OH OH
0
N)-LNH
O 0
0
H H
HO¨P¨O¨P¨O¨P-0
OH OH OH
OH OH
- 309 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
HO AN
1
0 0 0 0
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2 Hydroxymeth
N yl-CTP + 50
00901014005/ o o o t NO
% CTP/1- 0.4535 0.4546 0.4414
00901015002 HO-P-O-P-O-P-0
OH OH OH Methyl-
OH OH pseudo-UTP
0
N)-NH
O 0 00
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
HN)
N
1
0 0 0 N 0
II II II
HO-P-O-P-O-P-0 N4Ac-CTP/1-
OH OH OH
00901014007/ Methyl- 0.3213 0.2257 0.3696
00901015001 OH OH
pseudo-UTP
0
Th\l)-NH
O 0 00
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
HN)-
N
1
0 0 0 N 0
II II II
HO-P-O-P-O-P-0
OH OH OH
00901014007/ N4Ac-CTP/5-
0.2747 0.3903 0.2972 00901013007 OH OH Methoxy-UTP
0
0)-LNH
O 0 0 tNO
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 310 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 71. In vitro translation screen
The in vitro translation assay was done with the Rabbit Reticulocyte Lysate
(nuclease-
treated) kit (Promega, Madison, WI; Cat. # L4960) according to the
manufacturer's instructions. The
reaction buffer was a mixture of equal amounts of the amino acid stock
solutions devoid of Leucine or
Methionine provided in the kit. This resulted in a reaction mix containing
sufficient amounts of both
amino acids to allow effective in vitro translation.
The modRNAs of firefly Luciferase, human GCSF and human EPO, harboring
chemical
modifications on either the bases or the ribose units, were diluted in sterile
nuclease-free water to a
final concentration of 250ng in 2.5u1 (Stock 10Ong/p1). The modRNA (250ng) was
added to the
mixture of freshly prepared Rabbit Reticulocyte Lysate and reaction buffer.
The in vitro translation
reaction was done in a standard 0.2m1 polypropylene 96-well PCR plates (USA
Scientific, Ocala, FL;
Cat. # 1402-9596) at 30 C in a Thermocycler (MJ Research PCT-100, Watertown,
MA).
After 45min incubation, the reaction was stopped by placing the plate on ice.
Aliquots of
the in vitro translation reaction containing luciferase modRNA were
transferred to white opaque
polystyrene 96-well plates (Corning, Manassas, VA; Cat. # CL53912) and
combined with 100u1
complete luciferase assay solution (Promega, Madison, WI). The volumes of the
in vitro translation
reactions were adjusted or diluted until no more than 2 millon relative light
units per well were
detected for the strongest signal producing samples. The background signal of
the plates without
reagent was about 200 relative light units per well. The plate reader was a
BioTek Synergy H1
(BioTek, Winooski, VT).
Aliquots of the in vitro translation reaction containing human GCSF modRNA or
human
EPO mRNA were transferred and analyzed with a human GCSF-specific or EPO ELISA
kit (both from
R&D Systems, Minneapolis, MN; Cat. #s 5C550, DEPOO respectively) according to
the manufacturer
instructions. All samples were diluted until the determined values were within
the linear range of the
human GCSF or EPO ELISA standard curve.
- 311 -

Table 20: In vitro Translation Data.
Luc Epo
GCSF 0
Chemical Luc Std
Epo Std
Expression GSCF
n.)
Compound # Structure Expression
Expression
Modifications Dev
Dev Std Dev
(RLUs)
(pg/ml) (pg/ml)
vi
-1
oe
o vo
vi
1-,
1-,
HNNH
00902015001 o o s o PseudoU-alpha-
5221 480 2669
492 7763 538
(194)HO-
P-O-P-O-P-0 thio-TP
OH OH OH
OH OH
0
NANH
00902015002 o o s o 1-Methyl-
pseudo-U-alpha- 1201 840 1694
143 4244 44 P
(195) H H II
P-O-P-O-P-0
.
N,
HO-
OH OH OH
thio-TP
.
u,
.3
OH OH
No
0
1
0
0
0
1
,...----..NANH
r
0
03601015003 o o o o 1-Ethyl-pseudo-
122 36 7894
383 5700 288
(172)HO-
P-O-P-O-P-0 UTP
OH OH OH
OH OH
0
1\1)N1H
IV
03601015004 o o o o 1-Propyl-
140 7 838
36 1613 75
HO-
n
,-i
(173) H H H
P-O-P-O-P-0 pseudo-UTP
cp
OH OH OH ciL:)
n.)
o
1-,
OH OH
.6.
Ci3
-4
o
.6.
1-,
w
- 312 -

S
HN )-LNH
0
00901015006 0 o 00 2-Thio-pseudo-
(193) HO-P-O-P-O-P-0
UTP 2198 297 12310
2602 5988 238 n.)
=
1-,
OH OH OH
Ci3
oe
OH OH
vo
1¨,
1¨,
NH2
F3CN
00901014003 0 o o i 5-
II H H N 0 Trifluoromethyl- 23340 294 10200
817 31510 156
(351) HO-P-O-P-O-P-0
CTP
OH OH OH
OH OH
0
P
1 NH
N,
0
00901013004 N 0 o o 5-Methyl-2-thio-
i,
i,
(4) HO¨P¨O¨P¨O¨P-0
H H H
UTP 235 30 1100
11 2319 44
00
OH OH OH
"
0
r
0
'
OH OH

0
1
r
0
NH2
HON
00901014005 0 o o t NO 5-
H H H Hydroxymethyl- 154000 5090 9425
442 26600 462
(350) HO-P-O-P-O-P-0
CTP
OH OH OH
OH OH
IV
0 0
n
NH
1-3
00901013004 0 o o t NO UTP-5-oxyacetic
CP
n.)
(348) HO¨P¨O¨P¨O¨P-0
II H H
acid Me ester 162 30 544
32 4388 775
1-,
4=,
0
OH OH OH
=-.1
0
OH OH
.6.
1¨,
w
- 313 -

0
0
0 0 0 t N0t=.)
00901013007 II II II 5-methoxy-UTP 306600
619 17530 3678 26440 344 =
HO-P-O-P-O-P-0
OH OH OH
Ci3
oe
vo
OH OH
1-,
1-,
0
H11)-
N
00901014007 o r\l
o o 1 _L N4-Ac-CTP 167600 1461 8675 1790 8794
131
II H H " -ICI
HO-P-O-P-O-P-0
OH OH OH
OH OH
P
NH2
.
N,
BrN
0
w
03601014008 o o o
1 w
u,
II II II N 0 5-Bromo-CTP 194900 5665 8581
143 13510 1706 3
(379) HO-P-O-P-O-P-0
"
0
OH OH OH
r
0
1
0
0
OH OH
'
r
0
1\11112
H2N N
03601014009 o o o t N0 5-Aminoallyl-
II H H 887 242 169
3 1806 181
(381) HO-P-O-P-O-P-0 CTP
OH OH OH
OH OH
'V
n
c 4
=
. 6 .
- 4
=
. 6 .
- 314 -

0
_t
n.)
0 o 0
N N NH2 2-Aminopurine-
107000 28420
1025
22180
362 8675 1-,
03601012004 HO-P-O-P-O-P-0
vi
(382) riboside TP
OH OH OH
o
OH OH
1-,
1-,
0
)-NH
1
3744
244
0 0 0 S 2-Thio-UTP 1181 222 1894
92
00901013008 ii ii ii
HO-P-O-P-O-P-0
OH OH OH VL)
OH OH
P
O
N)
BrNH
w
w
u,
1
21530
1231 .
.3
0 0 0 N o 5-Bromo-UTP 218500 11290
18220 6
r.,
00901013009 ii ii ii
0
HO-P-O-P-O-P-0
r
0
OH OH OH
'
0
0
1
OH OH
r
0
1\11112
N
0 0 S
00902014001 ii ii ii NO Alpha-thio-CTP 142900
20660 17000 1671 26930 281
HO-P-O-P-O-P-0
OH OH OH
OH OH
'A
,¨i
0
cp
,..,
H2NNH
1
0 0 0 N 0
5-Aminoallyl-
14870 2587 1863
54 3706 706 o
1-,
.6.
00901013010 ii ii ii
-1
HO-P-O-P-O-P-0 UTP
-4
OH OH OH
o
.6.
1-,
w
OH OH
- 315 -

0
)11F1
O 0 S tN0
0
k.)
00902013001 Alpha-thio-UTP 51180 4835
14260 1465 20530 1381 =
II II II
HO-P-O-P-O-P-0
OH OH OH
Ci3
oe
o
OH OH
1-,
1-,
NH2
F3CN
0 0
0 1
N 0
ii II H
HO-P-O-P-O-P-0
OH OH OH 5-
00901014003/ OH OH Trifluoromethyl-
281
00901015002 o CTP/1-Methyl-
Th1)-LNH pseudo-UTP
P
o o o o
.
N)
HO-P-O-P-O-P-0
w
w
u,
OH OH OH
0
.3
N,
0
OH OH
r
0
1
NH2
0
0
1
HON
r
0
0 0 0 tNO
HO-P-O-P-O-P-0
OH OH OH 5-
00901014005/ OH OH Hydroxymethyl-
706
00901015002 o CTP/1-Methyl-
NANH pseudo-UTP
o o o o
IV
n
HO-P-O-P-O-P-0
1-3
OH OH OH
CP
t=.)
OH OH
=
1-,
.6.
Ci3
-4
o
.6.
1-,
w
- 316 -

NH2
Br
'N
0 0 0 tN
0
HO-P-O-P-O-P-0
H H H
t,.)
=
I..,
CA
OH OH OH
oe
03601014008/ OH OH 5-Bromo-CTP/1-
vD
vi
Methyl-pseudo-
1381
00901015002 o UTP
1¨,
NANH
0 0 0 o
HO-P-O-P-O-P-0
H H H
OH OH OH
OH OH
Table 21: In vitro Translation Data.
P
.
,,
In Vitro
In Vitro In Vitro
Translation Translation Translation
.3
Structure Chemical
,,
Luc
hEpo hGCSF .
Modifications
,
,
Expression Expression Expression
.
,
(RLUs)
(pg/ml) (pg/ml) ,
.
NH2 NH2
Br
'N N
0 0 0 1 1
N 0 0 0 0 N 0
HO-Fi'-0-Fi'-0-c'-0
HO-P-O-P-O-P-0
H H H
OH OH OH 1,c2_ OH OH OH 75% 5-Bromo-CTP
OH OH OH OH 25'Y CTP
372909 36208 21330 1-d
n
O 1-Methyl-pseudo-UTP
N)-LNH
CP
0 0 0
w
HO-P-O-P-O-P-0
II II II
I..,
.6,
-a
OH OH OH
-4
=
.66
OH OH
w
- 317 -

NH2 NH2
Br.,..1..,
0
'N
o
0 0 0
'''N 0 0 0 0
''''N 0
ii ii 1 1
CA
HO-P-O-P-O-P-0 HO-P-O-P-O-P-0
-O5
OH OH OH I:) OH OH OH
I:) oe
75% 5-Bromo-CTP
,.z
CA
OH OH OH OH 25% CTP 863775
24515 14760 1-

Pseudo-UTP
0
HNANH
0 0 0 "0
ii II H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2 NH2
Br.,..-I,
P
'N N
0
1 1
0 0 0N)'''N NO 0 0 0 N 0
ii II H
LoW'
HO-P-O-P-O-P-0
HO-P-O-P-O-P-0 u,
OH OH OH I:) OH OH OH ' ()
50% 5-Bromo-CTP
c,"
OH OH OH OH 50% CTP
o
1593328 33896 32040 ,
a,
ol
Pseudo-UTP
T
HNANH
0 0 0 "0
ii II H
HO-P-O-P-O-P-0
OH OH OH '1 IcL)
OH OH
IV
n
cp
k...)
=
.6.
--c-,.5
-4
=
.6.
,..,
- 318 -

NH2
Br NH2
N N
0 0 0 1 0 0 0
1 0
I\J 0
w
I\J 0
o
II II H
HO-P-O-P-O-P-0 H H H
HO-P-O-P-O-P-0
OH OH OH OH OH OH 1,c2_
cii
-a,
25% 5-Bromo-CTP
00
OH OH OH OH
o
75'Y CTP 193009
43143 63360 vi
1-

o Pseudo-UTP
HNANH
0 0 0 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2 0
F3CN OANH
P
o o o N(:)
0 0 0 1 ,L 5-Trifluoromethyl-CTP
43541
29120 115470 .
"
I\J 0 5-Methoxy-UTP
.
H H H
HO-P-O-P-O-P-0 H H H
HO-P-O-P-O-P-0
w
u,
OH OH OH OH OH OH
0
0
N,
OH OH OH OH
'
r
0
1
0
NH 0
m
I
HON OANH
0
0 0 0 1 1 5-Hydroxymethyl-CTP
N 0 0 0 0 121836
18398 26595
N 0 5-Methoxy-UTP
II II H
HO-P-O-P-O-P-0 ii ii ii
HO-P-O-P-O-P-0
OH OH OH OH OH OH
OH OH OH OH
NH2 0
BrN OANH
IV
0 0 0 1 1 5-Bromo-CTP
n
N 0 0 0 0 83463
23204 12330
II II H
HO-P-O-P-O-P-0 II N 0 II II
HO-P-O-P-O-P-0 5-Methoxy-UTP
OH OH OH OH OH OH
CP
w
o
OH OH OH OH
.6.
-a
-4
.6.
- 319 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 72. Transfection in HeLa Cells
The day before transfection, 20.000 HeLa cells (ATCC no. CCL-2; Manassas, VA)
were
harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand
Island, NY) and seeded
in a total volume of 100u1 EMEM medium (supplemented with 10%FCS and lx
Glutamax) per well in
a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at
37 C in 5% CO2
atmosphere overnight. Next day, 83ng of Luciferase modRNA or 250ng of human
GCSF modRNA,
harboring chemical modifications on the bases or the ribose units, were
diluted in lOul final volume of
OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000
(LifeTechnologies, Grand
Island, NY) was used as transfection reagent and 0.2plwere diluted in lOul
final volume of OPTI-
MEM. After 5min incubation at room temperature, both solutions were combined
and incubated
additional 15min at room temperature. Then the 20plwere added to the 100u1
cell culture medium
containing the HeLa cells. The plates were then incubated as described before.
After 18h to 22h incubation, cells expressing luciferase were lysed with 100p1
Passive
Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions.
Aliquots of the lysates
were transferred to white opaque polystyrene 96-well plates (Corning,
Manassas, VA) and combined
with 100u1 complete luciferase assay solution (Promega, Madison, WI). The
lysate volumes were
adjusted or diluted until no more than 2 mio relative light units per well
were detected for the strongest
signal producing samples. The background signal of the plates without reagent
was about 200
relative light units per well. The plate reader was a BioTek Synergy H1
(BioTek, Winooski, VT).
After 18h to 22h incubation, cell culture supernatants of cells expressing
human GCSF or
human EPO were collected and centrifuged at 10.00Orcf for 2min. The cleared
supernatants were
transferred and analyzed with a human GCSF-specific or EPO ELISA kit (both
from R&D Systems,
Minneapolis, MN; Cat. #s SCS50, DEPOO, respectively) according to the
manufacturer instructions.
All samples were diluted until the determined values were within the linear
range of the human GCSF
or EPO ELISA standard curve.
- 320 -

Table 22: HeLa Cell Transfection Data.
GCSF
Epo
0
Luc
Expressio
Chemical Luc Std
Expression Epo Std GSCF k.)
Compound # Structure Expression
n o
Modifications Dev
Dev Std Dev 1-,
(RLUs)
vi
(pg/ml)
-1
(pg/ml)
oe
o
vi
o 1-,
1-,
HNNH
00902015001 0 0 S "0 PseudoU-alpha-
(194) H H H
HO-P-O-P-O-P-0 thio-TP 2015 131.5
302800 2544 320000 1687
OH OH OH
OH OH
0
N)-
NH
00902015002 0 0 S "0
1-Methyl- P
H
pse udo-U-alpha- 4900 325.3
348600 7151 372100 4637
(195)
H H HO-P-O-P-O-P-0 r.,
'
thio-TP
OH OH
.3
OH OH
N,
0
r
0
1
0
0
0
,
r
,...----.NANH .
03601015003 0 0 0 "0 1-Ethyl-pseudo-
(172) H H H
HO-P-O-P-O-P-0 UTP 130.50 34.65
52780 1491 209300 3033
OH OH OH
OH OH
0
1\1)1\JH
n
03601015004 0 0 0 "0
1-Propyl- 1-3
(173) H H H
II
pseudo-UTP
0.00 0.00 10000 74.07
cp
n.)
OH OH OH ciL:)
o
1-,
.6.
OH OH
Ci3
-4
o
.6.
1-,
w
- 321 -

S
HN )-LNH
0
00901015006 o o HO- o o 2-Thio-pseudo-
1999 384.7
380600 4607 239300 10490 n.)
=
(193)
P-O-P-O-P-0 UTP
1-,
vi
OH OH OH
Ci3
oe
o
OH OH
1-,
1-,
NH2
F3CN
00901014003 o o o 1 5-
N 0 Trifluoromethyl- 32250 808.9
668100 2155 1039000 9891
HO-
(351) II H H
P-O-P-O-P-0
CTP
OH OH OH
OH OH
0
P
NH
0
"
00901013004 o o
5-Methyl-2-thio-
u,
N s
8333 57.47 16420 0.00
HO-
.
(4) H H H
P-O-P-O-P-0 UTP
3
r.,
OH OH OH

r
0
1
OH OH
'
0
1
r
0
Hrr
00901014004 o o 0 Cy
N N4-methyl CTP
90280 885.1
HO-
(346) H H H
P-O-P-O-P-0
OH OH OH
OH OH
IV
n
NH2
HON
5-
00901014005 o o HO- 1
N 0
o
Hydroxymethyl- 22160 754.5
440300 1931 1151000 39860 cp
n.)
=
(350) ii II H
P-O-P-O-P-0
O1-,
.6.
OH OH OH CTP
Ci3
-4
o
OH OH
.6.
1-,
w
- 322 -

0 0
OONH
0
00901013004 o o 0 NL0 UTP-5-oxyacetic
II H H
8333 402.3 5714 221.5
(348) HO-P-O-P-O-P-0 acid Me ester
OH OH OH
OH OH
0
0
NH
00901013005 5-Methoxy
O o o 0 N(:)
(358) carbonyl methyl-
8333 172.4
OH OH OH UTP
ciL:)
OH OH
0
0)-LNH
0
0 0 0
00901013007 II II II NO 5-methoxy-UTP
31580 1241 1116000 18170 1399000 2004
HO-P-O-P-O-P-0
OH OH OH
0
OH OH
0
0
HN
00901014007 o 0 o NO N4-Ac-CTP 141300 4463
2907000 41750 2094000 6826
II H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
¨
c
-:-
- 323 -

NH2
BrN
0
t0 N
t=.)
03601014008 o o
5-Bromo-CTP 125700 28470
3263000 42000 1003000 2605
(379)
=
1-,
II H H
HO-P-O-P-O-P-0
Ci3
OH OH OH
oe
o
OH OH
1-,
NH2
H2N N
1
0
03601014009 o o N 0 5-Aminoallyl-
319.00 8.49
3488 23.41 24290 571.43
HO-
(381) II H H
P-O-P-O-P-0 CTP
OH OH OH
OH OH
P
N--,----,--,N
2'
0 0 0 1
0
w
w
u,
N 'N NH2 2-Aminopurine-
.
713.50 3.54
56980 292.2 39290 205.7
03601012004 HO-P-O-P-O-P-0
.3
(382)
OH OH OH 1c2_ riboside TP
"
.
,
,
OH OH
0
0
1
r
0
0
)-L NH
t
00901013008 ii ii ii N
0 0 0 2-Thio-UTP 423.00 16.97
182600 1808 214300 4915
HO-P-O-P-O-P-0
OH OH OH
OH OH
IV
n
O
Br).
NH
CP
tt=.)
o
0 0 0 N
5-Bromo-UTP 2731 36.06
210500 3218 118600 3926
00901013009
.6.
II II II
HO-P-O-P-O-P-0
Ci3
OH OH OH
-4
o
.6.
1-,
OH OH
w
- 324 -

NH2
N
0
O 0 S NO
t=.)
00902014001 Alpha-thio-CTP 1845 1.41
195400 3733 285000.00 6925 =
II II II
HO-P-O-P-O-P-0
I..,
UI
OH OH OH
Ci3
oe
o
OH OH
1-,
1-,
0
...---.õ---,j1-..
H2N NH
O 0 0 1
5-Aminoallyl-
00901013010 0 1946 63.64
67440 1984 40710 211.0
II II II
HO-P-O-P-O-P-0
UTP
OH OH OH
OH OH
0
P
ANN
0
"
O 0 S 1
0
w
w
u,
00902013001 N o Alpha-thio-UTP 937.0 57.98 190700 8612
73570 923.5 .
HO-P-O-P-O-P-0
II II II
a'
IV
OH OH OH
0
r
0
1
OH OH

0
1
r
0
NH2
F3CN
0 0 0 tNO
HO-P-O-P-O-P-0
ii II H
OH OH OH 5-
00901014003/ OH OH Trifluoromethyl-
1668 254.6
492.2 2750 427100 5002
00901015002 o CTP/1-Methyl-
NANH pseudo-UTP
IV
n
1-i
O 0 00
HO-P-O-P-O-P-0
ii ii ii
CP
t=.)
o
OH OH OH
1-,
.6.
OH OH
Ci3
-4
o
.6.
1-,
w
- 325 -

NH2
HON
0 0 0
NO
0
HO-P-O-P-O-P-0
t=.)
=
1-,
OH OH OH
5-
-1
oe
00901014005/ OH OH Hydroxymethyl-
vo
22530 349.3
164800 17320 1666000 23170 vi
00901015002 o CTP/1-Methyl-
1¨,
1¨,
NANH pseudo-UTP
o o o o
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
BrN
0 0 0 NO
P
.
HO-P-O-P-O-P-0
H H
u,
OH OH OH
w
u,
0
03601014008/ 5-Bromo-CTP/1-
OH OH
.3
"
Methyl-pseudo- 28210 420.70
1248000 21190 474300 4124 0
,
00901015002 o UTP
cn
1
N)-LNH
0
0
1
r
0 0 0 o
0
HO-P-O-P-O-P-0
H H H
OH OH OH
OH OH
IV
n
c 4
c : ,
. 6 .
- 4
c : ,
. 6 .
- 326 -

NH2
F3C
'N
O 0 0
1 0
Thl
II II II 0
t=.)
HO-P-O-P-O-P-0
=
1-,
OH OH OH
5-
-1
oe
00901014003/ OH OH Trifluoromethyl-
vo
1340 231.2
429900 879 431400 4013 vi
00901015001 o CTP/Pseudo-
HN
1¨,
1¨,
ANH UTP
O 0 0 o
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
Br
'N
O 0 0
N P
HO-P-O-P-O-P-0
II II II
0
N,
0
OH OH OH
w
w
u,
0
03601014008/ OH OH 5-Bromo-
,,,
oo
CTP/Pseudo- 19340 224.9
859700 2097 355700 14150 0
00901015001 o UTP
,
,
,D
HNNH
1
O 0 0
o r
0
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
IV
n
,-i
cp
t..)
=
.6.
-,i-:--,
-4
=
.6.
- 327 -

NH2
F3C,,..}N
0 0 0 1
0
t=.)
II II II N 0
HO-P-O-P-O-P-0
o
1-,
OH OH OH
Ci3
oe
OH OH
o
1-,
1-,
NH2
' N75 % 5-
Trifluoromethyl-
00901014003/ tNO
0 0 o CTP + 25 % 1086 166.2
577900 4792 754300
00901015002 ii ii ii
HO-P-O-P-O-P-0 CTP/1-Methyl-
OH OH OH pseudo-UTP
OH OH
0
N)-
NH
P
N)
O o o o
'
II II II
HO-P-O-P-O-P-0
w
u,
0
0
OH OH OH kL0_
N,
0
r
OH OH
0
1
0
0
1
r
0
IV
n
1-i
cp
t.)
,-,
.6.
-,i-:--,
-4
.6.
,-,
- 328 -

NH2
F3C
'N
0 0 0 1
II II II N 0
0
HO-P-O-P-O-P-0
k.)
o
1-,
OH OH OH
Ci3
oe
OH OH
o
1-,
1-,
NH2 50 % 5-
N Trifluoromethyl-
00901014003/ t NO CTP + 50 % 3932 89.09
1043000 20620 1267000 8739
00901015002 o o 0
HO-P-O-P-O-P-0
H II H CTP/1-Methyl-
pseudo-UTP
OH OH OH
OH OH
0
NANH
P
O o o o
.
"
HO-P-O-P-O-P-0
w
w
u,
OH OH OH
0
00
1.,
OH OH
0
r
0
1
0
0
1
r
0
IV
n
1-i
cp
t.)
,-,
.6.
-,i-:--,
-4
.6.
,-,
- 329 -

NH2
F3C.,..}..N
1
o 0 0 N 0
0
t.)
o
II II II
HO-P-O-P-O-P-0
OH OH OH
Ci3
oe
o
OH OH
1-,
1-,
NH2
25 % 5-
N
00901014003/ o o o 1
N 0 Trifluoromethyl-
CTP + 75 % 15190 159.1
1991000 38850 2271000
00901015002 HO-P-O-P-O-P-0 CTP/1-Methyl-
OH OH OH pseudo-UTP
OH OH
0
NANH
00 0 o
P
. N,
II II II
HO-P-O-P-O-P-0
0
w
OH OH OH
w
u,
2
OH OH
N,
0
1
0
0
1
r
0
IV
n
1-i
cp
t.)
,-,
.6.
-,i-:--,
-4
.6.
,-,
- 330 -

NH2
Br
'N
0 0 0 1 N 0
0
HO-P-O-P-O-P-0
II II II
t=.)
o
1-,
OH OH OH cIL:)
cii
Ci3
oe
OH OH
o
1-,
1-,
NH2
N 50 % 5-Bromo-
03601014008/ tN0
o o o CTP + 50 %
45140 2274
2114000 59190 1921000 14350
00901015002
HO-P-O-P-O-P-0
CTP/1-Methyl-
pseudo-UTP
OH OH OH
OH OH
0
NANH
P
O o o o
o
r.,
HO-P-O-P-O-P-0
II II II
0
w
w
OH OH OH
u,
0
0
OH OH
N,
0
1
0
0
1
r
0
IV
n
1-i
cp
t.)
,-,
.6.
-,i-:--,
-4
.6.
,-,
- 331 -

NH2
Br
N
00 0
II II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
25 % 5-Bromo-
03601014008/CTP + 75 %
00901015002ii ii ii 76360 175.4
2782000 2903 2717000 4819
o o 0 tN,0 CTP/1-Methyl-
HO-P-O-P-O-P-0 pseudo-UTP
OH OH OH
OH OH
0
N)-LNH
O o 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
0
-a
- 332 -

NH2
HON
0
0 0 0 1
t=.)
o
II II II N 0
1-,
HO-P-O-P-O-P-0
OH OH OH
Ci3
oe
o
OH OH
1-,
1-,
NH2
50% 5-
00901014005/
1,[1 Hydroxymethyl-
o o o
00901015002 N 0 OTP + 50 % 54390 628.6
2307000 23850 2624000 25380
ii ii ii
HO-P-O-P-O-P-0 CTP/1-Methyl-
OH OH OH pseudo-UTP
OH OH
0
Th\l)-NH
P
oo o o
II II II
o
r.,
HO-P-O-P-O-P-0
.
w
OH OH OH
w
u,
0
0
OH OH
N,
0
1
0
'
0
HN A
1
r
0
N
0 0 0 tNO
ii II II
HO-P-O-P-O-P-0
OH OH OH (cL) N4Ac-CTP/1-
00901014007/
00901015002 OH OH Methyl-pseudo- 112200 633.6
2005000 4713 2074000 52510
UTP
o
NA
NH
IV
n
,-i
00 0 0
ii I, I,
cp
HO-P-O-P-O-P-0
t=.)
OH OH OH
o
1-,
.6.
OH OH
Ci3
-4
o
.6.
1-,
w
- 333 -

0
HN)
0
N
N
0
0 0 0 tN
H H H
HO-P-O-P-O-P-0
pe
O O O
yD
00901014007/ H H H N4Ac-CTP/5-
00901013007 OH OH Methoxy-UTP 7990
2724 420800 24440 611400 2199
o
o,)-L
NH
0 0 0 N
H II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
P
.
Table 23: HeLa Cell Transfection Data.
N)
GCSF
.
.3
Epo
Expressi N)Chemical GSCF Std 0
,
Structure Std on
.
Modifications
Dev
Dev
.
,
,
(pg/ml)
.
NH2 NH2
BrN N
0 0 0 1 1
0
0 0 II 1\1 0 1\1 0
HO-P-O-P-O-P-0 H H H
HO-P-O-P-O-P-0
OH OH OH L:34 OH OH OH c_ 4
OH OH OH OH 75% 5-Bromo-CTP
25'Y CTP
534332 2294872 1510500 1-d
o
Pseudo-UTP n
1\1ANH
1-3
0 0 0 0
CP
N
HO-P-O-P-O-P-0
=
1-,
OH OH OH
-a
-4
OH OH
o
.6.
1-,
W
- 334 -

NH2 NH2
BrAN
N
1 1
0
0 0 0 ''''N NO 0 0 0 ^ 0
N
ii II H
=
HO-P-O-P-O-P-0 HO-P-O-P-O-P-0
1-,
OH OH OH ' '''':) OH OH OH ' ''.:)
Un
75% 5-Bromo-CTP
oe
OH OH OH OH
25% CTP 729830
1650000 882500 vi

o
Pseudo-UTP 1¨
HN ANH
0 0 0
0
ii II H
HO-P-O-P-O-P-0
OH OH OH 'ThcL)
OH OH
NH2 NH2
13r.,..-1z,,
1 1 N
0 0
P
0 --..N 0 0 0 0 1
N 0
ii II H
II II H
HO-P-O-P-O-P-OHO-P-O-P-O-P-0
2
OH OH OH ' '''':) OH OH OH ' ''VL7
50% 5-Bromo-CTP
u,
o,
OH OH OH OH 102350
03
50 /0 CTP
1442308 1486000 n,
4
o
Pseudo-UTP ,E!
o,
ol
HN ANH
o,
,
0 0 0
18
0
1 1 II H
HO-P-O-P-O-P-0
OH OH OH' (1)
OH OH
IV
n
c 4
w
=
. 6 .
=
. 6 .
- 335 -

NH2 NH2
Br
' N N
O 0 0 1
1 0
1\1 0 0 0 0 Th \I 0
N
II II II
HO-P-O-P-O-P-0 H H H
HO-P-O-P-O-P-0
0
OH OH OH OH OH OH
OH OH OH OH 25% 5-Bromo-CTP
c,e
75'Y CTP 153026
1358974 1801500 vi

o Pseudo-UTP

HNANH
0 0 0 o
II II II
HO-P-O-P -0-P-0
OH OH OH
OH OH
NH2 0
F3C (:)j-L NH
' N
O 0 0 1 1
5-Trifluoromethyl-CTP P
N 0 0 0 0 1\1 0 16168
67949 351500 .
ii ii ii
HO-P-O-P-O-P-0 H H H
HO-P-O-P -0-P-0 5-Methoxy-UTP
"
OH OH OH k2_ OH OH OH
w
w
u,
0,
OH OH OH OH
00
IV
0
I
NH2 0
0
01
I
HON (:)-L NH
r
0
O 0 0 1 1 5-Hydroxymethyl-CTP
N 0 0 0 0 N 0 152072
921795 1361500
II II II
HO-P-O-P-O-P-0 H II II
HO-P-O-P -0-P-0 5-Methoxy-UTP
OH OH OH OH OH OH
OH OH OH OH
NH2 0
BrN 0j-L NH
IV
O 0 0 1 1
5-Bromo-CTP n
N 0 0 0 0
1-3
N 0 61114
951282 338500
II II II
HO-P-O-P-O-P-0 H H H
HO-P-O-P -0-P-0 5-Methoxy-UTP
cp
OH OH OH 1c24 OH OH OH
0
1-,
OH OH OH OH
.6.
-a
-4
.6.
- 336 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
Example 73. PBMC Cytokine Assay
A. PBMC isolation and Culture
50 mL of human blood from three donors was received from Research Blood
Components
(Brighton, MA) in sodium heparin tubes. For each donor, the blood was pooled
and diluted to 70 mL
with DPBS (Life Technologies, Grand Island, NY, 14190-250) and split evenly
between two 50 mL
conical tubes. 10 mL of Ficoll Paque (GE Healthcare, Fairfield, CT, 17-5442-
03) was gently
dispensed below the blood layer. The tubes were centrifuged at 2000 rpm for 30
minutes with low
acceleration and braking (Thermo, Waltham, MA, 75004506). The tubes were
removed and the buffy
coat PBMC layers were gently transferred to a fresh 50 mL conical and washed
with DPBS. The
tubes were centrifuged at 1450 rpm for 10 minutes.
The supernatant was aspirated and the PBMC pellets were resuspended and washed
in
50 mL of DPBS. The tubes were centrifuged at 1450 rpm for 10 minutes. This
wash step was
repeated, and the PBMC pellets were resuspended in 5 mL of OptiMEM
(LifeTechnologies,
31985088) and counted. The cell suspensions were adjusted to a concentration
of 3.0 x 106 cells /
mL live cells.
These cells were then plated on 96 well tissue culture treated round bottom
plates
(Corning Costar, Tewksbury MA, 3799) per donor at 50 pL per well. Within 30
minutes, transfection
mixtures were added to each well at a volume of 50 pL per well.
B. Trans fection Preparation
Modified mRNA encoding firefly Luciferase (mRNA SEQ ID NO: 4), human G-CSF
(mRNA
sequence shown in SEQ ID NO: 2; polyA tail of approximately 140 nucleotides
not shown in
sequence; 5'cap, Cap1) or human EPO (mRNA sequence shown in SEQ ID NO: 6;
polyA tail of
approximately 140 nucleotides not shown in sequence; 5'cap, Cap1) were diluted
to 100 ng / pL in a
final volume of 30 pL of sterile water.
Separately, for each mRNA sample, 2.4 pL of Lipofectamine 2000
(LifeTechnologies
11668019) was diluted with 268 pL OptiMEM. In a 96 well plate the aliquots of
30 pL of each mRNA
was added to 270.4 pL of the diluted Lipofectamine 2000. The plate containing
the mRNA to be
transfected was incubated for 20 minutes. The transfection mixtures were then
transferred to each of
the human PBMC plates at 50 pL per well (6 wells per mRNA sample). The plates
were then
incubated at 37 C. After 2 hours incubation, 11p1 of heat-inactivated FCS
(LifeTechnologies,
16140071) was added to each well (10 /OFCS final concentration).
The plates were further incubated at 37 C, 5 % CO2 for additional 18-20hs. In
order to
harvest the supernatant, plates were centrifuged at 1450rpm for 5min in a
swinging plate rotor. The
supernatant of 6 wells transfected with the same mRNA was carefully harvested
and pooled in a
single well of a fresh 96-well plate. Supernatants were either frozen or used
fresh until ELISA
analysis was done.
- 337 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
Innate Immune Response Analysis
The ability of unmodified and modified mRNA to trigger innate immune
recognition as
measured by interferon-alpha production. Use of in vitro PBMC cultures is an
accepted way to
determine the immunostimulatory potential of oligonucleotides (Robbins et al.,
Oligonucleotides 2009
19:89-102). The release of interferon was measured with an IFN-alpha multi-
subtype ELISA (PBL
interferonsource, Pisctaway, NJ, 11668019) following the instructions of the
manufacturer.
Table 24: PBMC Assay Data.
Luc hEPO
hGCSF
Chemical (3 Donor (3 Donor
(3 Donor
Compound # Structure
Modifications samples) samples)
samples)
pg/ml pg/ml
pg/ml
o
HN ANH
00902015001"o PseudoU-alpha- 20 -190 50
(194) 9 9
I'-0-I'-0-I'-0 thio-TP 170 75
508.33
HO-F F F
-90 400
640
OH OH OH L:D4
OH OH
0
N ANN
00902015002 9 "o 1-Methyl- 180 -290
425
(195) 9
HO-1-0-1-0-1-0 pseudo-U-alpha- 500 512.5
916.66
OH OH OH thio-TP 180 475 1250
OH OH
S
HN ANH
009010150069 "o 2-Thio-pseudo- 530 -210
358.33
(193) 9 9 1-0-1-0-1-0 UTP 1180 675
166.66
HO-
OH OH OH
1400 362.5
490
LC)4
OH OH
NH2
F3C
00901016002 1 1 5- 6670 4440
6253.33
N 0 Trifluoromethyl- 2190
3100 6725
HO-
(351) 9 9 9
1-0-1-0-1-0
OH OH OH CTP 6410 1412.5 6280
OH OH
NH2
HON 4960
00901014005 9 9 1 ,L 5- 7130 3100
6575
(350) 9
HO-1-0-1-0-1-0
OH OH OH N 0 Hydroxymethyl- 3680 2412.5
5800
CTP 8990
8180
OH OH
- 338 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
o
C))-LN H -210
O o o t N 390
162.5
350
00901013007 ii ii ii 5-methoxy-UTP -70
-41.66
HO-P-O-P-O-P-0 150
OH OH OH -170 40
OH OH
0
H11)-
N 7170 4050 5683.33
00901014007 o o o 1 _L N4-Ac-CTP 2500 2137.5
4883.33
ii II H r\l" -10 5
HO-P-O-P-O-P-0 879 5850 4590
OH OH OH
OH OH
NH2
BrN
03601014008 o o o 1 _L 5470 2300
2808.33
ii II H - -C, 5-Bromo-CTP 1080 487.5
2266.67
(379) HO-P-O-P-O-P-0
5420 500 1650
OH OH OH kL0_
OH OH
NH2
F3CN
0 0 0 N
HO-P-O-P-O-P-0
OH OH OH k 0
-13
5- -184
-61 61.11
00901014003/ OH OH Trifluoromethyl- 25
-121 13.88
00901015002 o CTP/1-Methyl-
Thl)-LNH pseudo-UTP
o
H II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
HON
0 0 0 1
N 0
HO-P-O-P-O-P-0
OH OH OH 5- 135
00901014005/ OH OH Hydroxymethyl- 775
762 3
108.33
00901015002 o CTP/1-Methyl-
1163 3 13.88
ANH pseudo-UTP
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 339 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
Br
1 1
9 9 9 N 0
HO-PI-0-PI-0-PI-0
OH OH OH 13
03601014008/ OH OH 5-Bromo-CTP/1- -178
-33 -27.77
Methyl-pseudo- -140 -
36.11
00901015002 -27
o UTP 27.77
N)-LNH
o
9 9 9
HO-1-0-1-0-1-0
OH OH OH VL)
OH OH
NH2
F3C
1 1
9 9 9 N 0
HO-1-0-1-0-1-0
OH OH OH 5- 118.75 97
00901014003/ OH OH Trifluoromethyl- 237.5 12
102.77
00901015001 o CTP/Pseudo- 186.1 30
111.11
HN
ANH UTP
o
9 9 9
HO-1-0-1-0-1-0
OH OH OH
OH OH
NH2
Br
1 1
9 9 9 N 0
HO-PI-0-PI-0--0
OH OH OH 165
513.8
9
03601014008/ OH OH 5-Bromo- 1296
CTP/Pseudo- 706.2
280.5
00901015001 o UTP 800 12
HN)-LNH
o
9 9 9
HO-1-0-1-0-1-0
OH OH OH
OH OH
- 340 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
NO
0 0 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
75 % 5-
-181.25 -100
Trifluoromethyl- _206.25 -58 -
19.44
CTP + 25 %
0 -64 -
213.8
00901014003/
0 0 9
00901015002 o
HO-P-O-P-O-P-0 CTP/1-Methyl-
pseudo-UTP
OH OH OH
OH OH
0
11)-LNH
0 0 9
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
0 0 9 -N NO
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2 50 % 5- 37.5 -52
-55.55
Trifluoromethyl- _193.75 -70
00901015002 0 0 9
00901014003/ NO CTP + 50 %
5.555 -130 -
47.22
CTP/1-Methyl-
HO-P-O-P-O-P-0
pseudo-UTP
OH OH OH
OH OH
0
N)-LNH
0 0 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 341 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
0 0 0 0
H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
25 % 5-
1006
00901014003/ o o oN Trifluoromethyl-
CTP + 75 % 1175 216
39
41.66
00901015002 HO-P-O-P-O-P-0 CTP/1-Methyl- 663.8
-79 -
19.44
OH OH OH pseudo-UTP
OH OH
0
ANH
0 0
0
H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
BrN
0 0 0 0
H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
50 % 5-Bromo-
03601014008/
O o o NO CTP + 50% 200 -39 27.77
190.6 -130 -36.11
CTP/1-Methyl-
00901015002
HO-P-O-P-O-P-0 50
pseudo-UTP
OH OH OH
OH OH
0
ANH
0 0
0
H H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 342 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
BrN
0 N
t 0 0
H II II
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
/1\1 25 % 5-Bromo-
318.7 148
03601014008/ 1 _{, CTP + 75 %
-58 -
166.6
O o 0 -73 -8.333446.8
II II II
00901015002 N- CTP/1-Methyl-
130.5
HO-P-O-P-O-P-0 pseudo-UTP
OH OH OH
OH OH
0
NANH
0 0
0 o
H H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
HO 'N
1
0 0 0 N 0
H II H
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH2
806
tNHydroxymethyl- 1650
115
580.55
00901014005/ o o 0 CTP + 50 % 1370
00901015002
HO-P-O-P-O-P-0 CTP/1-Methyl- 752.7
224 83.33
OH OH OH pseudo-UTP
OH OH
0
N)-LNH
0 0
0 o
II H H
HO-P-O-P-O-P-0
OH OH OH
OH OH
0
A
HAI
1 II
0 0 0 N 0
ii II H
HO-P-O-P-O-P-0
OH OH OH
00901014007/ N4Ac-CTP/1- -96 -77
-19.44
Methyl-pseudo- -65 -136 -36.11
OH OH
00901015002
UTP -33 -136
o
N)-LNH
0 0 0 o
HO-P-O-P-O-P-0
OH OH OH
OH OH
- 343 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
0
HN A
----tr''N
0 0
0 1
N 0
HO-P11-041-041-0
OH OH OH .1 <--) -171.8 -87
00901014007/ N4Ac-CTP/5- -84 -155 8.333
00901013007 OH OH Methoxy-UTP -75 -155 30.55
o
OANH
0 0
0 1
H H H N 0
HO-P-O-P-O-P-0
OH OH OH
OH OH
NH: NH:
N
? o, 'i 'fli"...0 0 0 0 ,,,,..0
HO -1--0 N7-0 -0 H0 4-04,34_,,B6
OH OH OH 'IcO4 OH OH OH 75% 5-Bromo-
-19 -38 56
OH OH OH OH CTP
03601014008/ -80 56 78
25% CTP
00901015002 'N1NH -33 78 -4
1-Methyl-
0 0 0 :0
HO 4-0 -P-0 -0-0 pseudo-UTP
OH OH OH
OH OH
NH, NH,
el
81
,1)l
O
HO -13-0 -13-C4-0 'N ICg)ICg)3 N C
. HO--O--O-0 . -0 H OH OH -6 OH OH OH .6 75% 5-Bromo- -2
33 85
OH OH OH OH
03601014008/ CTP -145 85
120
00901015001 HNINH 25% CTP 33 120 1
HO -1'3-0 -10
-10
-0 L'''.--- Pseudo-UTP
6H 6H 6H -v.L_D
OH OH
NH2 NH2
a a a p 0 CB:21c
HO -49-04-0-0-0 H0-4?., -0 0 -11.-0.,14
OH OH OH . OH OH OH 50% 5-Bromo- 102 56 76
OH OH OH OH
03601014008/ CTP -135 76 92
00901015001 HNINH 50% CTP 56 92 44
1,-11-01
H0 0 -0, j Pseudo-UTP
OH OH OH 0
OH OH
NH, NH,
Br N'el N
HO LO LO LO N HO -I: -0 -?'.-0 4-0
OH OH OH -6 OH OH OH ----6 25% 5-Bromo- -34 -18
OH OH OH OH 149
03601014008/ CTP -135 149
213
00901015001 HN:LNH 75% CTP -18 213
420
0 0 0 ,..--..--0 Pseudo-UTP
H04-04-04-0
OH OH OH
OH OH
NH2 0
0 0 0 F3C to 0 0
0 I -. 5- -169 -72 41
00901014003/ H04-04-0-0-0 HO -13-0-0-04-0
Trifluoromethyl- -170 41 39
OH OH OH OH OH OH
00901013007 CTP -72 39 27
OH OH OH OH
5-Methoxy-UTP
- 344 -

CA 02933568 2016-06-10
WO 2015/089511 PCT/US2014/070413
NH2
HO --el ,c).& 5- -176 -116 36
00901014005/ HO -ILO -13-0 HO -ILO -1?'-0 Hydroxymethyl-
-140 36 109
6H 6H 6H 6H 6H 6H
00901013007 CTP -116 109 -8
OH OH OH OH
5-Methoxy-UTP
NH2 0
Sr'e'1,1 -165 -111 27
03601014008/ H4-4-4¨ HO -12-0 -12-0 I 5-Bromo-CTP -
197 -27 88
00901013007 OH OH OH
OH OH OH OH
OH OH 5-Methoxy-UTP -111 88 -6
Example 74. Cytokine screen of modRNA with novel chemistries in BJ Fibroblast
cells
At 2 or 3 days prior to transfection, 100,000 BJ fibroblast cells (ATCC no.
CRL-2522; Manassas, VA)
were harvested by treatment with trypsin-EDTA solution (LifeTechnologies,
Grand Island, NY) and
seeded in a total volume of 500 ul EMEM medium (supplemented with 10 /OFCS and
10% Glutamax,
both LifeTechnologies, Grand Island, NY) per well in 24-well cell culture
plates (Corning, Manassas,
VA). The cells were grown at 37 C in a 5% CO2 atmosphere overnight. On the
next day, 500 ng
modRNA, harboring chemical modifications on the bases or the ribose units,
were diluted in 25 ul final
volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000
(LifeTechnologies,
Grand Island, NY) was used as transfection reagent and 1.0 ul was diluted in
25 ul final volume of
OPTI-MEM. After 5 min incubation at room temperature, both solutions were
combined and incubated
an additional 15 min at room temperature. The 50 ul were added to the 500 ul
cell culture medium
containing the BJ fibroblast cells. The plates were then incubated as
described above.
After 18 h to 22 h incubation, cell culture supernatants were collected and
centrifuged at 10,000 rcf
for 2 min. The cleared supernatants were transferred and analyzed with a human
IFN-beta ELISA
(R&D Systems, Minneapolis, MN; Cat. #s 41410-2) and human CCL-5/RANTES ELISA
(R&D
Systems, Minneapolis, MN; Cat. #s SRNOOB) according to the manufacturer
instructions. All samples
were diluted until the determined values were within the linear range of the
ELISA standard curves
using a BioTek Synergy H1 plate reader (BioTek, Winooski, VT).
Table 25: Cytokine screen results in BJ Fibroblast cells.
hGCSF
hEpo mRNA
Luc mRNA mRNA
mRNA Chemistry RANTES
RANTES [pg/m1] RANTES
[pg/m1]
[pg/m1]
N4-acetyl-cytidine TP, ATP, GTP,
2546 4360 4103
UTP
5-methoxy-uridine TP, ATP, GTP,
33.33 -6.66 -6.66
UTP
- 345 -

CA 02933568 2016-06-10
WO 2015/089511
PCT/US2014/070413
pseudouridine TP, ATP, GTP,
4600 5490 5016
CTP
1-methyl-pseudouridine TP, ATP,
5473 8780 4816
GTP, CTP
2-thio-pseudouridine TP, ATP,
1706 5440 2106
GTP, CTP
5-hydroxymethyl-cytidine TP,
9826 2160 9063
ATP, GTP, UTP
5-bromocytidine TP, ATP, GTP,
1380 1343 1900
UTP
5-trifluromethylcytidine TP, ATP,
2303 7593 4203
GTP, UTP
OTHER EMBODIMENTS
It is to be understood that while the present disclosure has been described in
conjunction with the
detailed description thereof, the foregoing description is intended to
illustrate and not limit the scope
of the present disclosure, which is defined by the scope of the appended
claims. Other aspects,
advantages, and modifications are within the scope of the following claims.
- 346 -

Representative Drawing

Sorry, the representative drawing for patent document number 2933568 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Time Limit for Reversal Expired 2019-12-17
Application Not Reinstated by Deadline 2019-12-17
Letter Sent 2019-12-16
Letter Sent 2019-12-16
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2018-12-17
Letter Sent 2016-09-08
Inactive: Single transfer 2016-09-02
Inactive: Sequence listing - Amendment 2016-08-19
BSL Verified - No Defects 2016-08-19
Inactive: Sequence listing - Received 2016-08-19
Letter Sent 2016-07-28
Letter Sent 2016-07-28
Letter Sent 2016-07-28
Letter Sent 2016-07-28
Inactive: Single transfer 2016-07-22
IInactive: Courtesy letter - PCT 2016-07-21
BSL Verified - No Defects 2016-07-20
Inactive: Sequence listing - Amendment 2016-07-20
Inactive: Sequence listing - Received 2016-07-20
Inactive: Cover page published 2016-07-08
Inactive: Notice - National entry - No RFE 2016-06-29
Inactive: First IPC assigned 2016-06-22
Inactive: IPC assigned 2016-06-22
Inactive: IPC assigned 2016-06-22
Inactive: IPC assigned 2016-06-22
Application Received - PCT 2016-06-22
Inactive: Sequence listing - Received 2016-06-10
BSL Verified - Defect(s) 2016-06-10
National Entry Requirements Determined Compliant 2016-06-10
Inactive: Court decision rendered 2016-01-02
Application Published (Open to Public Inspection) 2015-06-18

Abandonment History

Abandonment Date Reason Reinstatement Date
2018-12-17

Maintenance Fee

The last payment was received on 2017-11-20

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2016-06-10
Registration of a document 2016-07-22
Registration of a document 2016-09-02
MF (application, 2nd anniv.) - standard 02 2016-12-15 2016-11-22
MF (application, 3rd anniv.) - standard 03 2017-12-15 2017-11-20
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MODERNA THERAPEUTICS, INC.
Past Owners on Record
ANDREW W. FRALEY
ANTONIN DE FOUGEROLLES
ATANU ROY
CHRISTOPHER R. CONLEE
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2016-06-09 346 15,060
Claims 2016-06-09 17 890
Abstract 2016-06-09 1 55
Cover Page 2016-07-07 1 25
Notice of National Entry 2016-06-28 1 195
Reminder of maintenance fee due 2016-08-15 1 112
Courtesy - Certificate of registration (related document(s)) 2016-07-27 1 104
Courtesy - Certificate of registration (related document(s)) 2016-07-27 1 104
Courtesy - Certificate of registration (related document(s)) 2016-07-27 1 104
Courtesy - Certificate of registration (related document(s)) 2016-07-27 1 104
Courtesy - Certificate of registration (related document(s)) 2016-09-07 1 102
Courtesy - Abandonment Letter (Maintenance Fee) 2019-01-27 1 174
Reminder - Request for Examination 2019-08-18 1 117
Commissioner's Notice: Request for Examination Not Made 2020-01-05 1 537
Commissioner's Notice - Maintenance Fee for a Patent Application Not Paid 2020-01-26 1 534
International search report 2016-06-09 2 91
National entry request 2016-06-09 2 78
Declaration 2016-06-09 1 27
Sequence listing - Amendment 2016-07-19 1 26
Correspondence 2016-07-20 2 59
Sequence listing - Amendment 2016-08-18 2 64

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :